1
|
Wen T, Liu T, Chen H, Liu Q, Shen X, Hu Q. Demethylzeylasteral alleviates inflammation and colitis via dual suppression of NF-κB and STAT3/5 by targeting IKKα/β and JAK2. Int Immunopharmacol 2024; 142:113260. [PMID: 39340986 DOI: 10.1016/j.intimp.2024.113260] [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: 07/15/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is a common inflammatory bowel disease and a risk factor of colorectal cancer. Demethylzeylasteral (DZT), a bioactive component mainly isolated from Tripterygium wilfordii, has been shown to inhibit inflammation and cancer. However, its anti-UC function and molecular mechanisms have not been well characterized. This study aims to explore the therapeutic effect and functional targets of demethylzeylasteral against UC. METHODS RT-qPCR, Western blot and ELISA were used to detect the generation of pro-inflammatory cytokines and chemokines in murine macrophage cells. Luciferase reporter gene, Western blot, pull-down, CETSA, DARTS, and virtual docking were employed to detect the anti-inflammatory targets and molecular mechanisms of demethylzeylasteral. The anti-inflammatory and anti-colitis effects of demethylzeylasteral were further determined in DSS-challenged mice. RESULTS In vitro, demethylzeylasteral inhibited NO and PGE2 production by suppressing the mRNA and protein expression of iNOS and COX-2, and suppressed the mRNA expression of TNF-α, IL-1β, IL-6, MCP-1, CXCL9, and CXCL10 in RAW264.7 macrophages stimulated by LPS/IFNγ. Furthermore, demethylzeylasteral was not only capable of inhibiting IKKα/β-NF-κB activation, but also able to block JAKs-STAT3/5 activation in LPS/INFγ-incubated RAW264.7 cells or DSS-exposed colon tissues of mice. Mechanistically, demethylzeylasteral was found to directly bind to IKKα/β and JAK2 kinases, leading to inactivation of pro-inflammatory signaling cascades and reduced generation of cytokines and chemokines. In vivo, oral administration of demethylzeylasteral significantly attenuated DSS-induced colitis, which was mainly manifested as mitigated symptoms of colitis, colonic mucosal barrier damage, and colonic inflammation. CONCLUSION We demonstrated that demethylzeylasteral alleviated UC pathology by blocking NF-κB and STAT3/5 pathways via targeting IKKα/β and JAK2 kinases, raising the possibility that demethylzeylasteral could act as a candidate for the treatment of UC.
Collapse
Affiliation(s)
- Tian Wen
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Ting Liu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Hongqing Chen
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qi Liu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Xiaofei Shen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Qiongying Hu
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| |
Collapse
|
2
|
Yang L, Ren Q, Wang Y, Zheng Y, Du F, Wang F, Zhou J, Gui L, Chen S, Chen X, Zhang W, Sun Y, Zhong X, Liu H, Jiang X, Zhang Z. Research progress of mitochondrial dysfunction induced pyroptosis in acute lung injury. Respir Res 2024; 25:398. [PMID: 39511593 DOI: 10.1186/s12931-024-03028-1] [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: 04/01/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024] Open
Abstract
Acute lung injury (ALI) is a common critical respiratory disease in clinical practice, especially in the ICU, with a high mortality rate. The pathogenesis of ALI is relatively complex, mainly involving inflammatory response imbalance, oxidative stress, cell apoptosis, and other aspects. However, currently, the treatment measures taken based on the above mechanisms have not had significant effects. Recent research shows that mitochondrial dysfunction and pyroptosis play an important role in ALI, but there is not much analysis on the relationship between mitochondrial dysfunction and pyroptosis at present. This article reviews the situation of mitochondrial dysfunction in ALI, pyroptosis in ALI, whether mitochondrial dysfunction is related to pyroptosis in ALI, and how to do so, and further analyzes the relationship between them in ALI. This review describes how to alleviate mitochondrial dysfunction, and then suppress the associated immunological pyroptosis, providing new ideas for the clinical treatment of ALI.
Collapse
Affiliation(s)
- Luhan Yang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Qingyi Ren
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yaohui Wang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yucheng Zheng
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Fei Du
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Fang Wang
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jie Zhou
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Linxi Gui
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Shengdong Chen
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiurui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Wanting Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yuhong Sun
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiaolin Zhong
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Hao Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| | - Xian Jiang
- Department of Anesthesiology, Luzhou People's Hospital, Luzhou, 646000, China.
| | - Zhuo Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
3
|
Merchak AR, Bolen ML, Tansey MG, Menees KB. Thinking outside the brain: Gut microbiome influence on innate immunity within neurodegenerative disease. Neurotherapeutics 2024:e00476. [PMID: 39482179 DOI: 10.1016/j.neurot.2024.e00476] [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: 08/19/2024] [Revised: 10/18/2024] [Accepted: 10/19/2024] [Indexed: 11/03/2024] Open
Abstract
The complex network of factors that contribute to neurodegeneration have hampered the discovery of effective preventative measures. While much work has focused on brain-first therapeutics, it is becoming evident that physiological changes outside of the brain are the best target for early interventions. Specifically, myeloid cells, including peripheral macrophages and microglia, are a sensitive population of cells whose activity can directly impact neuronal health. Myeloid cell activity includes cytokine production, migration, debris clearance, and phagocytosis. Environmental measures that can modulate these activities range from toxin exposure to diet. However, one of the most influential mediators of myeloid fitness is the gut microenvironment. Here, we review the current data about the role of myeloid cells in gastrointestinal disorders, Parkinson's disease, dementia, and multiple sclerosis. We then delve into the gut microbiota modulating therapies available and clinical evidence for their use in neurodegeneration. Modulating lifestyle and environmental mediators of inflammation are one of the most promising interventions for neurodegeneration and a systematic and concerted effort to examine these factors in healthy aging is the next frontier.
Collapse
Affiliation(s)
- Andrea R Merchak
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - MacKenzie L Bolen
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Malú Gámez Tansey
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA; Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| | - Kelly B Menees
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
4
|
Liu X, Dong Y, Wang C, Guo Z. Application of chitosan as nano carrier in the treatment of inflammatory bowel disease. Int J Biol Macromol 2024; 278:134899. [PMID: 39187100 DOI: 10.1016/j.ijbiomac.2024.134899] [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: 12/11/2023] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024]
Abstract
Inflammatory bowel disease (IBD), encompassing ulcerative colitis (UC) and Crohn's disease (CD), is characterized by persistent and recurrent gastrointestinal inflammation. Conventional IBD therapies often involve the use of antibiotics, NSAIDs, biological agents, and immunomodulators. While these medications can mitigate acute inflammatory symptoms, their long-term efficacy is frequently compromised due to cumulative toxic effects. In recent years, significant attention has shifted toward nanoparticle (NP)-based therapies as potential alternatives for IBD management. Various drug delivery strategies, including those targeting microbiota interactions, ligand-receptor binding, pH sensitivity, biodegradability, pressure response, and specific charge and size parameters, have been explored and optimized in animal studies. This review provides a comprehensive overview of the current landscape of chitosan NP-mediated drug delivery systems for IBD treatment. Additionally, it will discuss the prevailing challenges and propose future research directions to advance chitosan NP-based therapeutic strategies for IBD.
Collapse
Affiliation(s)
- Xiaoming Liu
- Department of Gastroenterology, Huaihe Hospital of Henan University, 115 Ximen Street, Kaifeng 475000, Henan, China
| | - Yunrui Dong
- Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, China
| | - Chenyu Wang
- Department of General Surgery, Huaihe Hospital of Henan University, 115 Ximen Street, Kaifeng 475000, Henan, China
| | - Zhiguo Guo
- Department of Gastroenterology, Suzhou Hospital of Anhui Medical University (Suzhou Municipal Hospital of Anhui Province), No.616 Bianyangsan Road, Suzhou 234000, Anhui, China.
| |
Collapse
|
5
|
Duan KL, Wang TX, You JW, Wang HN, Wang ZQ, Huang ZX, Zhang JY, Sun YP, Xiong Y, Guan KL, Ye D, Chen L, Liu R, Yuan HX. PCK2 maintains intestinal homeostasis and prevents colitis by protecting antibody-secreting cells from oxidative stress. Immunology 2024; 173:339-359. [PMID: 38934051 DOI: 10.1111/imm.13827] [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: 02/06/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Maintaining intracellular redox balance is essential for the survival, antibody secretion, and mucosal immune homeostasis of immunoglobulin A (IgA) antibody-secreting cells (ASCs). However, the relationship between mitochondrial metabolic enzymes and the redox balance in ASCs has yet to be comprehensively studied. Our study unveils the pivotal role of mitochondrial enzyme PCK2 in regulating ASCs' redox balance and intestinal homeostasis. We discover that PCK2 loss, whether globally or in B cells, exacerbates dextran sodium sulphate (DSS)-induced colitis due to increased IgA ASC cell death and diminished antibody production. Mechanistically, the absence of PCK2 diverts glutamine into the TCA cycle, leading to heightened TCA flux and excessive mitochondrial reactive oxygen species (mtROS) production. In addition, PCK2 loss reduces glutamine availability for glutathione (GSH) synthesis, resulting in a decrease of total glutathione level. The elevated mtROS and reduced GSH expose ASCs to overwhelming oxidative stress, culminating in cell apoptosis. Crucially, we found that the mitochondria-targeted antioxidant Mitoquinone (Mito-Q) can mitigate the detrimental effects of PCK2 deficiency in IgA ASCs, thereby alleviating colitis in mice. Our findings highlight PCK2 as a key player in IgA ASC survival and provide a potential new target for colitis treatment.
Collapse
Affiliation(s)
- Kun-Long Duan
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Tian-Xiang Wang
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jian-Wei You
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Hai-Ning Wang
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhi-Qiang Wang
- Department of Immunology, School of Basic Medical Sciences, Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zi-Xuan Huang
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jin-Ye Zhang
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yi-Ping Sun
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yue Xiong
- Cullgen Inc., San Diego, California, USA
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Dan Ye
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital of Fudan University, Key Laboratory of Metabolism and Molecular Medicine (Ministry of Education), Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Li Chen
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Ronghua Liu
- Shanghai Fifth People's Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hai-Xin Yuan
- Shanghai Fifth People's Hospital, Molecular and Cell Biology Research Lab of Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Lv Y, Peng J, Ma X, Liang Z, Salekdeh GH, Ke Q, Shen W, Yan Z, Li H, Wang S, Ding X. Network Analysis of Gut Microbial Communities Reveals Key Reason for Quercetin Protects against Colitis. Microorganisms 2024; 12:1973. [PMID: 39458282 PMCID: PMC11509604 DOI: 10.3390/microorganisms12101973] [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: 07/31/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
As one of the most representative natural products among flavonoids, quercetin (QUE) has been reported to exhibit beneficial effects on gut health in recent years. In this study, we utilized a dextran sulfate sodium (DSS)-induced colitis mice model to explore the protective effects and underlying mechanisms of QUE on colitis. Our data demonstrated that QUE oral gavage administration significantly ameliorates the symptoms and histopathological changes associated with colitis. Additionally, the concentration of mucin-2, the number of goblet cells, and the expression of tight junction proteins (such as ZO-1, Occludin, and Claudin-1) were all found to be increased. Furthermore, QUE treatment regulated the levels of inflammatory cytokines and macrophage polarization, as well as the oxidative stress-related pathway (Nrf2/HO-1) and associated enzymes. Additionally, 16S rDNA sequencing revealed that QUE treatment rebalances the alterations in colon microbiota composition (inlcuding Bacteroidaceae, Bacteroides, and Odoribacter) in DSS-induced colitis mice. The analysis of network dynamics reveals a significant correlation between gut microbial communities and microenvironmental factors associated with inflammation and oxidative stress, in conjunction with the previously mentioned findings. Collectively, our results suggest that QUE has the potential to treat colitis by maintaining the mucosal barrier, modulating inflammation, and reducing oxidation stress, which may depend on the reversal of gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Yanan Lv
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Jing Peng
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Xiaoyu Ma
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010010, China
| | - Zeyi Liang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Ghasem Hosseini Salekdeh
- Department of Systems Biology, Agricultural Biotechnology Research Institute of Iran, Agricultural Research, Education, and Extension Organization, Karaj 3135933151, Iran;
- Department of Molecular Sciences, Macquarie University, North Ryde, NSW 2109, Australia
| | - Qunhua Ke
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Wenxiang Shen
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Zuoting Yan
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Hongsheng Li
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Shengyi Wang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| | - Xuezhi Ding
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou 730050, China; (Y.L.); (J.P.); (X.M.); (Z.L.); (Q.K.); (W.S.); (Z.Y.); (H.L.)
| |
Collapse
|
7
|
Liu H, Guan L, Su X, Zhao L, Shu Q, Zhang J. A broken network of susceptibility genes in the monocytes of Crohn's disease patients. Life Sci Alliance 2024; 7:e202302394. [PMID: 38925865 PMCID: PMC11208737 DOI: 10.26508/lsa.202302394] [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: 09/26/2023] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Genome-wide association studies have identified over 200 genetic loci associated with inflammatory bowel disease; however, the mechanism of such a large amount of susceptibility genes remains uncertain. In this study, we integrated bioinformatics analysis and two independent single-cell transcriptome datasets to investigate the expression network of 232 susceptibility genes in Crohn's disease (CD) patients and healthy controls. The study revealed that most of the susceptibility genes are specifically and strictly expressed in the monocytes of the human intestinal tract. The susceptibility genes established a network within the monocytes of health control. The robustness of a gene network may prevent disease onset that is influenced by the genetic and environmental alteration in the expression of susceptibility genes. In contrast, we showed a sparse network in pediatric/adult CD patients, suggesting the broken network contributed to the CD etiology. The network status of susceptibility genes at the single-cell level of monocytes provided novel insight into the etiology.
Collapse
Affiliation(s)
- Hankui Liu
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Clin Lab, BGI Genomics, Shijiazhuang, China
- BGI Genomics, Shenzhen, China
| | - Liping Guan
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Clin Lab, BGI Genomics, Shijiazhuang, China
- BGI Genomics, Shenzhen, China
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Xi Su
- BGI Genomics, Shenzhen, China
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Lijian Zhao
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Clin Lab, BGI Genomics, Shijiazhuang, China
- BGI Genomics, Shenzhen, China
- Hebei Medical University, Shijiazhuang, China
| | - Qing Shu
- Department of Gastroenterology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Jianguo Zhang
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Clin Lab, BGI Genomics, Shijiazhuang, China
- BGI Research, Shenzhen, China
- Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
8
|
Argue BMR, Casten LG, McCool S, Alrfooh A, Gringer Richards J, Wemmie JA, Magnotta VA, Williams AJ, Michaelson J, Fiedorowicz JG, Scroggins SM, Gaine ME. Patterns of Immune Dysregulation in Bipolar Disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.26.24311078. [PMID: 39211848 PMCID: PMC11361205 DOI: 10.1101/2024.07.26.24311078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Bipolar disorder is a debilitating mood disorder associated with a high risk of suicide and characterized by immune dysregulation. In this study, we used a multi-faceted approach to better distinguish the pattern of dysregulation of immune profiles in individuals with BD. Methods We analyzed peripheral blood mononuclear cells (bipolar disorder N=39, control N=30), serum cytokines (bipolar disorder N=86, control N=58), whole blood RNA (bipolar disorder N=25, control N=25), and whole blood DNA (bipolar disorder N=104, control N=66) to identify immune-related differences in participants diagnosed with bipolar disorder compared to controls. Results Flow cytometry revealed a higher proportion of monocytes in participants with bipolar disorder together with a lower proportion of T helper cells. Additionally, the levels of 18 cytokines were significantly elevated, while two were reduced in participants with bipolar disorder. Most of the cytokines altered in individuals with bipolar disorder were proinflammatory. Forty-nine genes were differentially expressed in our bipolar disorder cohort and further analyses uncovered several immune-related pathways altered in these individuals. Genetic analysis indicated variants associated with inflammatory bowel disease also influences bipolar disorder risk. Discussion Our findings indicate a significant immune component to bipolar disorder pathophysiology and genetic overlap with inflammatory bowel disease. This comprehensive study supports existing literature, whilst also highlighting novel immune targets altered in individuals with bipolar disorder. Specifically, multiple lines of evidence indicate differences in the peripheral representation of monocytes and T cells are hallmarks of bipolar disorder.
Collapse
|
9
|
Dovrolis N, Valatas V, Drygiannakis I, Filidou E, Spathakis M, Kandilogiannakis L, Tarapatzi G, Arvanitidis K, Bamias G, Vradelis S, Manolopoulos VG, Paspaliaris V, Kolios G. Landscape of Interactions between Stromal and Myeloid Cells in Ileal Crohn's Disease; Indications of an Important Role for Fibroblast-Derived CCL-2. Biomedicines 2024; 12:1674. [PMID: 39200138 PMCID: PMC11351973 DOI: 10.3390/biomedicines12081674] [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: 06/06/2024] [Revised: 07/20/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND AND AIMS Monocyte recruitment in the lamina propria and inflammatory phenotype driven by the mucosal microenvironment is critical for the pathogenesis of inflammatory bowel disease. However, the stimuli responsible remain largely unknown. Recent works have focused on stromal cells, the main steady-state cellular component in tissue, as they produce pro-inflammatory chemokines that contribute to the treatment-resistant nature of IBD. METHODS We studied the regulation of these processes by examining the communication patterns between stromal and myeloid cells in ileal Crohn's disease (CD) using a complete single-cell whole tissue sequencing analysis pipeline and in vitro experimentation in mesenchymal cells. RESULTS We report expansion of S4 stromal cells and monocyte-like inflammatory macrophages in the inflamed mucosa and describe interactions that may establish sustained local inflammation. These include expression of CCL2 by S1 fibroblasts to recruit and retain monocytes and macrophages in the mucosa, where they receive signals for proliferation, survival, and differentiation to inflammatory macrophages from S4 stromal cells through molecules such as MIF, IFNγ, and FN1. The overexpression of CCL2 in ileal CD and its stromal origin was further demonstrated in vitro by cultured mesenchymal cells and intestinal organoids in the context of an inflammatory milieu. CONCLUSIONS Our findings outline an extensive cross-talk between stromal and myeloid cells, which may contribute to the onset and progression of inflammation in ileal Crohn's disease. Understanding the mechanisms underlying monocyte recruitment and polarization, as well as the role of stromal cells in sustaining inflammation, can provide new avenues for developing targeted therapies to treat IBD.
Collapse
Affiliation(s)
- Nikolas Dovrolis
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (M.S.); (L.K.); (G.T.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Vassilis Valatas
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (M.S.); (L.K.); (G.T.); (K.A.); (V.G.M.); (G.K.)
- Gastroenterology and Hepatology Research Laboratory, Medical School, University of Crete, 71003 Heraklion, Greece;
| | - Ioannis Drygiannakis
- Gastroenterology and Hepatology Research Laboratory, Medical School, University of Crete, 71003 Heraklion, Greece;
| | - Eirini Filidou
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (M.S.); (L.K.); (G.T.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Michail Spathakis
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (M.S.); (L.K.); (G.T.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Leonidas Kandilogiannakis
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (M.S.); (L.K.); (G.T.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Gesthimani Tarapatzi
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (M.S.); (L.K.); (G.T.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Konstantinos Arvanitidis
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (M.S.); (L.K.); (G.T.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Giorgos Bamias
- GI Unit, 3 Department of Internal Medicine, Sotiria Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Stergios Vradelis
- Second Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Vangelis G. Manolopoulos
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (M.S.); (L.K.); (G.T.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | | | - George Kolios
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (M.S.); (L.K.); (G.T.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| |
Collapse
|
10
|
Erdem I, Aktas S, Ogut S. Neohesperidin Dihydrochalcone Ameliorates Experimental Colitis via Anti-Inflammatory, Antioxidative, and Antiapoptosis Effects. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:15715-15724. [PMID: 38961631 DOI: 10.1021/acs.jafc.4c02731] [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: 07/05/2024]
Abstract
Neohesperidin dihydrochalcone (NHDC) is a citrus-originated, seminatural sweetener. There is no investigation concerning the effect of NHDC on ulcerative colitis. The purpose of this study was to determine the therapeutic and protective effects of NHDC in Wistar Albino rats. NHDC was given for 7 days after or before colitis induction. The results showed that NHDC significantly reduced the interleukin-6 (IL-6), interleukin-10 (IL-10), transforming growth factor-β1 (TGF-β1), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) levels. Catalase levels did not show a significant difference between the groups. NHDC provided a remarkable decrease in the expression levels of cyclooxygenase-2 (COX-2), myeloperoxidase (MPO), malondialdehyde (MDA), 8-hydroxy-2'-deoxyguanosine (8-OHdG), and nuclear factor kappa B (NF-κB). Total antioxidant status (TAS) levels were significantly elevated in NHDC treatment groups, while total oxidant status (TOS) and oxidative stress index (OSI) levels were significantly decreased. NHDC provided remarkable improvement in histological symptoms such as epithelial erosion, edema, mucosal necrosis, inflammatory cell infiltration, and hemorrhage. Also, caspase-3 expression levels were statistically decreased in NHDC treatment groups. The results indicated that NHDC might be a protection or alternative treatment for ulcerative colitis.
Collapse
Affiliation(s)
- Ilayda Erdem
- Department of Nutrition and Dietetics, Aydin Adnan Menderes University, Aydin 09010, Turkey
| | - Serdar Aktas
- Faculty of Veterinary Medicine, Aydin Adnan Menderes University, Aydin 09010, Turkey
| | - Serdal Ogut
- Department of Nutrition and Dietetics, Aydin Adnan Menderes University, Aydin 09010, Turkey
| |
Collapse
|
11
|
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
- 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
- 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
- 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
- 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
- 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
- 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
- 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
|
12
|
Zhang B, Sun C, Zhu Y, Qin H, Kong D, Zhang J, Shao B, Li X, Ren S, Wang H, Hao J, Wang H. Upregulation of TCPTP in Macrophages Is Involved in IL-35 Mediated Attenuation of Experimental Colitis. Mediators Inflamm 2024; 2024:3282679. [PMID: 38962170 PMCID: PMC11221972 DOI: 10.1155/2024/3282679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 04/11/2024] [Accepted: 06/01/2024] [Indexed: 07/05/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic intestinal inflammatory disease with complex etiology. Interleukin-35 (IL-35), as a cytokine with immunomodulatory function, has been shown to have therapeutic effects on UC, but its mechanism is not yet clear. Therefore, we constructed Pichia pastoris stably expressing IL-35 which enables the cytokines to reach the diseased mucosa, and explored whether upregulation of T-cell protein tyrosine phosphatase (TCPTP) in macrophages is involved in the mechanisms of IL-35-mediated attenuation of UC. After the successful construction of engineered bacteria expressing IL-35, a colitis model was successfully induced by giving BALB/c mice a solution containing 3% dextran sulfate sodium (DSS). Mice were treated with Pichia/IL-35, empty plasmid-transformed Pichia (Pichia/0), or PBS by gavage, respectively. The expression of TCPTP in macrophages (RAW264.7, BMDMs) and intestinal tissues after IL-35 treatment was detected. After administration of Pichia/IL-35, the mice showed significant improvement in weight loss, bloody stools, and shortened colon. Colon pathology also showed that the inflammatory condition of mice in the Pichia/IL-35 treatment group was alleviated. Notably, Pichia/IL-35 treatment not only increases local M2 macrophages but also decreases the expression of inflammatory cytokine IL-6 in the colon. With Pichia/IL-35 treatment, the proportion of M1 macrophages, Th17, and Th1 cells in mouse MLNs were markedly decreased, while Tregs were significantly increased. In vitro experiments, IL-35 significantly promoted the expression of TCPTP in macrophages stimulated with LPS. Similarly, the mice in the Pichia/IL-35 group also expressed more TCPTP than that of the untreated group and the Pichia/0 group.
Collapse
Affiliation(s)
- Baoren Zhang
- Department of General SurgeryTianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Chenglu Sun
- Department of General SurgeryTianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Yanglin Zhu
- Department of General SurgeryTianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Hong Qin
- Department of General SurgeryTianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Dejun Kong
- School of MedicineNankai University, Tianjin, China
| | - Jingyi Zhang
- Department of General SurgeryTianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Bo Shao
- Department of General SurgeryTianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Xiang Li
- Department of General SurgeryTianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Shaohua Ren
- Department of General SurgeryTianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Hongda Wang
- Department of General SurgeryTianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Jingpeng Hao
- Department of General SurgeryTianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
- Department of Anorectal SurgeryTianjin Medical University Second Hospital, Tianjin, China
| | - Hao Wang
- Department of General SurgeryTianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China
| |
Collapse
|
13
|
Zhong Z, Xu M, Ge C, Tan J. Exploring shared molecular signatures and regulatory mechanisms in nonalcoholic steatohepatitis and inflammatory bowel disease using integrative bioinformatics analysis. Sci Rep 2024; 14:12085. [PMID: 38802459 PMCID: PMC11130338 DOI: 10.1038/s41598-024-62310-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] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
The co-existence of inflammatory bowel disease (IBD) and non-alcoholic steatohepatitis (NASH) has raised interest in identifying shared molecular mechanisms and potential therapeutic targets. However, the relationship between these two diseases remains unclear and effective medical treatments are still lacking. Through the bioinformatics analysis in this study, 116 shared differentially expressed genes (SDEGs) were identified between IBD and NASH datasets. GO and KEGG pathway analyses revealed significant involvement of SDEGs in apoptotic processes, cell death, defense response, cytokine and chemokine activity, and signaling pathways. Furthermore, weighted gene co-expression network analysis (WGCNA) identified five shared signature genes associated specifically with IBD and NASH, they were CXCL9, GIMAP2, ADAMTS5, GRAP, and PRF1. These five genes represented potential diagnostic biomarkers for distinguishing patients with diseases from healthy individuals by using two classifier algorithms and were positively related to autophagy, ferroptosis, angiogenesis, and immune checkpoint factors in the two diseases. Additionally, single-cell analysis of IBD and NASH samples highlighted the expression of regulatory genes in various immune cell subtypes, emphasizing their significance in disease pathogenesis. Our work elucidated the shared signature genes and regulatory mechanisms of IBD and NASH, which could provide new potential therapies for patients with IBD and NASH.
Collapse
Affiliation(s)
- Zixuan Zhong
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, People's Republic of China.
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, People's Republic of China.
| | - Minxuan Xu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, People's Republic of China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, People's Republic of China
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, People's Republic of China
| | - Chenxu Ge
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, People's Republic of China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, People's Republic of China
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, People's Republic of China
| | - Jun Tan
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, People's Republic of China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, People's Republic of China
| |
Collapse
|
14
|
Liao X, Liu J, Guo X, Meng R, Zhang W, Zhou J, Xie X, Zhou H. Origin and Function of Monocytes in Inflammatory Bowel Disease. J Inflamm Res 2024; 17:2897-2914. [PMID: 38764499 PMCID: PMC11100499 DOI: 10.2147/jir.s450801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/23/2024] [Indexed: 05/21/2024] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), is a chronic disease resulting from the interaction of various factors such as social elements, autoimmunity, genetics, and gut microbiota. Alarmingly, recent epidemiological data points to a surging incidence of IBD, underscoring an urgent imperative: to delineate the intricate mechanisms driving its onset. Such insights are paramount, not only for enhancing our comprehension of IBD pathogenesis but also for refining diagnostic and therapeutic paradigms. Monocytes, significant immune cells derived from the bone marrow, serve as precursors to macrophages (Mφs) and dendritic cells (DCs) in the inflammatory response of IBD. Within the IBD milieu, their role is twofold. On the one hand, monocytes are instrumental in precipitating the disease's progression. On the other hand, their differentiated offsprings, namely moMφs and moDCs, are conspicuously mobilized at inflammatory foci, manifesting either pro-inflammatory or anti-inflammatory actions. The phenotypic spectrum of these effector cells, intriguingly, is modulated by variables such as host genetics and the subtleties of the prevailing inflammatory microenvironment. Notwithstanding their significance, a palpable dearth exists in the literature concerning the roles and mechanisms of monocytes in IBD pathogenesis. This review endeavors to bridge this knowledge gap. It offers an exhaustive exploration of monocytes' origin, their developmental trajectory, and their differentiation dynamics during IBD. Furthermore, it delves into the functional ramifications of monocytes and their differentiated progenies throughout IBD's course. Through this lens, we aspire to furnish novel perspectives into IBD's etiology and potential therapeutic strategies.
Collapse
Affiliation(s)
- Xiping Liao
- Clinical Medical Research Center, the Second Affiliated Hospital, Army Medical University, Chongqing, People’s Republic of China
- Department of Gastroenterology, the Second Affiliated Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Ji Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, People’s Republic of China
| | - Xiaolong Guo
- Clinical Medical Research Center, the Second Affiliated Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Ruiping Meng
- Clinical Medical Research Center, the Second Affiliated Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Wei Zhang
- Clinical Medical Research Center, the Second Affiliated Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Jianyun Zhou
- Clinical Medical Research Center, the Second Affiliated Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Xia Xie
- Clinical Medical Research Center, the Second Affiliated Hospital, Army Medical University, Chongqing, People’s Republic of China
- Department of Gastroenterology, the Second Affiliated Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Hongli Zhou
- Clinical Medical Research Center, the Second Affiliated Hospital, Army Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
15
|
Sharma J, Khan S, Singh NC, Sahu S, Raj D, Prakash S, Bandyopadhyay P, Sarkar K, Bhosale V, Chandra T, Kumaravelu J, Barthwal MK, Gupta SK, Srivastava M, Guha R, Ammanathan V, Ghoshal UC, Mitra K, Lahiri A. ORMDL3 regulates NLRP3 inflammasome activation by maintaining ER-mitochondria contacts in human macrophages and dictates ulcerative colitis patient outcome. J Biol Chem 2024; 300:107120. [PMID: 38417794 PMCID: PMC11065740 DOI: 10.1016/j.jbc.2024.107120] [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: 08/27/2023] [Revised: 02/06/2024] [Accepted: 02/20/2024] [Indexed: 03/01/2024] Open
Abstract
Genome-wide association studies in inflammatory bowel disease have identified risk loci in the orosomucoid-like protein 3/ORMDL sphingolipid biosynthesis regulator 3 (ORMDL3) gene to confer susceptibility to ulcerative colitis (UC), but the underlying functional relevance remains unexplored. Here, we found that a subpopulation of the UC patients who had higher disease activity shows enhanced expression of ORMDL3 compared to the patients with lower disease activity and the non-UC controls. We also found that the patients showing high ORMDL3 mRNA expression have elevated interleukin-1β cytokine levels indicating positive correlation. Further, knockdown of ORMDL3 in the human monocyte-derived macrophages resulted in significantly reduced interleukin-1β release. Mechanistically, we report for the first time that ORMDL3 contributes to a mounting inflammatory response via modulating mitochondrial morphology and activation of the NLRP3 inflammasome. Specifically, we observed an increased fragmentation of mitochondria and enhanced contacts with the endoplasmic reticulum (ER) during ORMDL3 over-expression, enabling efficient NLRP3 inflammasome activation. We show that ORMDL3 that was previously known to be localized in the ER also becomes localized to mitochondria-associated membranes and mitochondria during inflammatory conditions. Additionally, ORMDL3 interacts with mitochondrial dynamic regulating protein Fis-1 present in the mitochondria-associated membrane. Accordingly, knockdown of ORMDL3 in a dextran sodium sulfate -induced colitis mouse model showed reduced colitis severity. Taken together, we have uncovered a functional role for ORMDL3 in mounting inflammation during UC pathogenesis by modulating ER-mitochondrial contact and dynamics.
Collapse
Affiliation(s)
- Jyotsna Sharma
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shaziya Khan
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Nishakumari C Singh
- Sophisticated Analytical Instrument Facility and Research Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shikha Sahu
- Department of Gastroenterology, Sanjay Gandhi postgraduate institute of medical sciences, Lucknow, India
| | - Desh Raj
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shakti Prakash
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | | | - Kabita Sarkar
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Vivek Bhosale
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Tulika Chandra
- Department of Transfusion Medicine, Kings George Medical University, Lucknow, India
| | - Jagavelu Kumaravelu
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manoj Kumar Barthwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shashi Kumar Gupta
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Mrigank Srivastava
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India; Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rajdeep Guha
- Lab Animal Facility, CSIR-Central Drug Research Institute, Lucknow, India
| | - Veena Ammanathan
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Uday C Ghoshal
- Department of Gastroenterology, Sanjay Gandhi postgraduate institute of medical sciences, Lucknow, India
| | - Kalyan Mitra
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India; Sophisticated Analytical Instrument Facility and Research Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Amit Lahiri
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
16
|
Curci D, Franzin M, Zudeh G, Bramuzzo M, Lega S, Decorti G, Stocco G, Lucafò M. Expression profiles of the lncRNA antisense GAS5-AS1 in colon biopsies from pediatric inflammatory bowel disease patients and its role in regulating sense transcript GAS5. Eur J Pediatr 2024; 183:1657-1665. [PMID: 38197962 PMCID: PMC11001710 DOI: 10.1007/s00431-023-05403-4] [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: 10/12/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/11/2024]
Abstract
The long non-coding RNA (lncRNA) growth arrest-specific transcript 5 (GAS5) level was demonstrated as involved in pediatric inflammatory bowel disease (IBD) pathogenesis. Since its antisense transcript GAS5-AS1 has never been investigated in IBD, this study aims to detect whether GAS5-AS1 and GAS5 levels are related to IBD clinical parameters and investigate their correlation in vitro. Twenty-six IBD pediatric patients were enrolled; paired inflamed and non-inflamed intestinal biopsies were collected. We evaluated GAS5 and GAS5-AS1 levels by real-time PCR. The role of GAS5 and GAS5-AS1 was assessed in vitro by transient silencing in THP1-derived macrophages. GAS5-AS1 and GAS5 levels were associated with patients' clinical parameters; GAS5-AS1 expression was downregulated in inflamed tissues and inversely correlated with disease activity. A positive correlation between GAS5-AS1 and GAS5 levels was observed in non-inflamed biopsies. On THP1-derived macrophages, a reduced amount of both GAS5-AS1 and GAS5 was observed; accordingly, matrix metalloproteinase (MMP) 9 was increased. After GAS5-AS1 silencing, a downregulation of GAS5 was found, whereas no effect was detected on GAS5-AS1 after GAS5 silencing. Conclusion: This study provided for the first time new insights into the potential role of GAS5-AS1 in IBD. GAS5-AS1 modulates GAS5 levels in vitro and may serve as a potential IBD diagnostic biomarker. What is Known: • GAS5 is involved in regulating intestinal MMP-2 and MMP-9 in pediatric patients with IBD; • GAS5-AS1 has never been investigated in the context of IBD; • GAS5-AS1 regulates the expression of GAS5, increasing its stability in tissues and in vitro cell models of cancer. What is New: • GAS5-AS1 correlated with GAS5 and IBD clinical parameters; • GAS5-AS1 can modulate GAS5 levels in macrophages; • GAS5-AS1 may serve as potential IBD diagnostic biomarker.
Collapse
Affiliation(s)
- Debora Curci
- Department of Advanced Translational Diagnostics, Institute for Maternal and Child Health IRCCS "Burlo Garofolo", 34137, Trieste, Italy
| | - Martina Franzin
- Department of Advanced Translational Diagnostics, Institute for Maternal and Child Health IRCCS "Burlo Garofolo", 34137, Trieste, Italy
| | - Giulia Zudeh
- Department of Advanced Translational Diagnostics, Institute for Maternal and Child Health IRCCS "Burlo Garofolo", 34137, Trieste, Italy
| | - Matteo Bramuzzo
- Department of Pediatric Gastroenterology, Institute for Maternal and Child Health IRCCS "Burlo Garofolo", 34137, Trieste, Italy
| | - Sara Lega
- Department of Pediatric Gastroenterology, Institute for Maternal and Child Health IRCCS "Burlo Garofolo", 34137, Trieste, Italy
| | - Giuliana Decorti
- Department of Medicine Surgery and Health Sciences, University of Trieste, 34149, Trieste, Italy
| | - Gabriele Stocco
- Department of Advanced Translational Diagnostics, Institute for Maternal and Child Health IRCCS "Burlo Garofolo", 34137, Trieste, Italy.
- Department of Medicine Surgery and Health Sciences, University of Trieste, 34149, Trieste, Italy.
| | - Marianna Lucafò
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| |
Collapse
|
17
|
Dai C, Dong ZY, Wang YN, Huang YH, Jiang M. Development and validation of a nomogram to predict non-response to 5-aminosalicylic acid in patients with ulcerative colitis. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2024; 116:124-131. [PMID: 37706463 DOI: 10.17235/reed.2023.9703/2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
BACKGROUND there are some patients with ulcerative colitis (UC) who have non-response (NR) to 5-aminosalicylic acid (5-ASA). To promote individualized treatment in UC patients, it is crucial to identify valid predictors to estimate NR to 5-ASA. Therefore, this study aimed to identify the predictive value of clinical and biochemical markers and to construct a nomogram model predicting NR to 5-ASA in patients with UC. METHODS data of patients diagnosed with UC in the First Hospital of China Medical University between January 2012 and December 2020 were retrospectively analyzed. Primary outcome was the proportion of NR to 5-ASA. Multivariable logistic regression was used to construct prediction models. Area under the curve (AUC), calibration and decision curve analyses (DCA) were assessed in the validation cohort. RESULTS of 284 UC patients who were treatment-naive, 86 (30.3 %) had NR to 5-ASA. Univariate regression analysis showed that disease classification (DC) (p = 0.008), monocytes (MONO) (p = 0.041), platelet distribution width (PDW) (p = 0.027), serum total cholesterol (TC) (p = 0.031) and α1 globulin (p < 0.001) were strongly associated with NR to 5-ASA. Receiver operating characteristics (ROC) analysis indicated the AUC was 0.852, it showed that this model has a good degree of discrimination. The DCA curve showed that the predicted probability is 0.0-96.0 %. CONCLUSION this study developed a predictive model with good discrimination and calibration, and high clinical validity, which can effectively estimate the risk of NR to 5-ASA. DC, MONO, PDW, TC and α1 globulin can be used as predictors for NR to 5-ASA in UC patients.
Collapse
Affiliation(s)
- Cong Dai
- Gastroenterology, First Hospital of China Medical University, china
| | - Zi-Yuan Dong
- Gastroenterology, First Hospital of China Medical University
| | - Yi-Nuo Wang
- Gastroenterology, First Hospital of China Medical University
| | - Yu-Hong Huang
- Gastroenterology, First Hospital of China Medical University
| | - Min Jiang
- Gastroenterology, First Hospital of China Medical University
| |
Collapse
|
18
|
Dong K, Zhang Y, Ji HR, Guan ZL, Wang DY, Guo ZY, Deng SJ, He BY, Xing JF, You CY. Dexamethasone-Loaded Lipid Calcium Phosphate Nanoparticles Treat Experimental Colitis by Regulating Macrophage Polarization in Inflammatory Sites. Int J Nanomedicine 2024; 19:993-1016. [PMID: 38299194 PMCID: PMC10829593 DOI: 10.2147/ijn.s442369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/20/2024] [Indexed: 02/02/2024] Open
Abstract
Background The M1/M2 polarization of intestinal macrophages exerts an essential function in the pathogenesis of ulcerative colitis (UC), which can be adjusted to alleviate the UC symptoms. Purpose A kind of pH-sensitive lipid calcium phosphate core-shell nanoparticles (NPs), co-loading with dexamethasone (Dex) and its water-soluble salts, dexamethasone sodium phosphate (Dsp), was constructed to comprehensively regulate macrophages in different states towards the M2 phenotype to promote anti-inflammatory effects. Methods Dex and Dsp were loaded in the outer lipid shell and inner lipid calcium phosphate (Cap) core of the LdCaPd NPs, respectively. Then, the morphology of NPs and methods for determining drug concentration were investigated, followed by in vitro protein adsorption, stability, and release tests. Cell experiments evaluated the cytotoxicity, cellular uptake, and macrophage polarization induction ability of NPs. The in vivo distribution and anti-inflammatory effect of NPs were evaluated through a 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced BALB/c mice ulcerative colitis model. Results The LdCaPd NPs showed a particle size of about 200 nm and achieved considerable loading amounts of Dex and Dsp. The in vitro and in vivo studies revealed that in the acidic UC microenvironment, the cationic lipid shell of LdCaPd underwent protonated dissociation to release Dex first for creating a microenvironment conducive to M2 polarization. Then, the exposed CaP core was further engulfed by M1 macrophages to release Dsp to restrict the pro-inflammatory cytokines production by inhibiting the activation and function of the nuclear factor kappa-B (NF-κB) through activating the GC receptor and the NF kappa B inhibitor α (I-κBα), respectively, ultimately reversing the M1 polarization to promote the anti-inflammatory therapy. Conclusion The LdCaPd NPs accomplished the sequential release of Dex and Dsp to the UC site and the inflammatory M1 macrophages at this site, promoting the regulation of macrophage polarization to accelerate the remission of UC symptoms.
Collapse
Affiliation(s)
- Kai Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- School of Pharmacy, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Ying Zhang
- School of Pharmacy, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Hong Rui Ji
- School of Pharmacy, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Ze Lin Guan
- School of Pharmacy, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Dan Yang Wang
- School of Pharmacy, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Zi Yang Guo
- School of Pharmacy, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Shu Jing Deng
- School of Pharmacy, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Bin Yang He
- School of Pharmacy, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Jian Feng Xing
- School of Pharmacy, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Cui Yu You
- Department of Pharmacy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| |
Collapse
|
19
|
Zeng Z, Cheng S, Li X, Liu H, Lin J, Liang Z, Liu X, Cao C, Li S, He X, Kang L, Wu X, Zheng X. Glycolytic Activation of CD14+ Intestinal Macrophages Contributes to the Inflammatory Responses via Exosomal Membrane Tumor Necrosis Factor in Crohn's Disease. Inflamm Bowel Dis 2024; 30:90-102. [PMID: 37406645 DOI: 10.1093/ibd/izad117] [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: 11/23/2022] [Indexed: 07/07/2023]
Abstract
BACKGROUND Macrophage (Mφ) activation plays a critical role in the inflammatory response. Activated Mφ go through profound reprogramming of cellular metabolism. However, changes in their intracellular energy metabolism and its effect on inflammatory responses in Crohn's disease (CD) remain currently unclear. The aim of this study is to explore metabolic signatures of CD14+ Mφ and their potential role in CD pathogenesis as well as the underlying mechanisms. METHODS CD14+ Mφ were isolated from peripheral blood or intestinal tissues of CD patients and control subjects. Real-time flux measurements and enzyme-linked immunosorbent assay were used to determine the inflammatory states of Mφ and metabolic signatures. Multiple metabolic routes were suppressed to determine their relevance to cytokine production. RESULTS Intestinal CD14+ Mφ in CD patients exhibited activated glycolysis compared with those in control patients. Specifically, macrophagic glycolysis in CD largely induced inflammatory cytokine release. The intestinal inflammatory microenvironment in CD elicited abnormal glycolysis in Mφ. Mechanistically, CD14+ Mφ derived exosomes expressed membrane tumor necrosis factor (TNF), which engaged TNFR2 and triggered glycolytic activation via TNF/nuclear factor κB autocrine and paracrine signaling. Importantly, clinically applicable anti-TNF antibodies effectively prevented exosomal membrane TNF-induced glycolytic activation in CD14+ Mφ. CONCLUSIONS CD14+ Mφ take part in CD pathogenesis by inducing glycolytic activation via membrane TNF-mediated exosomal autocrine and paracrine signaling. These results provide novel insights into pathogenesis of CD and enhance understanding of the mechanisms of anti-TNF agents.
Collapse
Affiliation(s)
- Ziwei Zeng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of General Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sijing Cheng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xuanna Li
- Department of Residents Standarization Training, Perking University Shenzhen Hospital, Shenzhen, China
| | - Huashan Liu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinxin Lin
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhenxing Liang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xuanhui Liu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chao Cao
- Center for Human Nutrition, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Shujuan Li
- Department of Pharmacy, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaowen He
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liang Kang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaojian Wu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaobin Zheng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
20
|
Yu S, Guo H, Ji Z, Zheng Y, Wang B, Chen Q, Tang H, Yuan B. Sea Cucumber Peptides Ameliorate DSS-Induced Ulcerative Colitis: The Role of the Gut Microbiota, the Intestinal Barrier, and Macrophage Polarization. Nutrients 2023; 15:4813. [PMID: 38004208 PMCID: PMC10674221 DOI: 10.3390/nu15224813] [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: 10/27/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
The incidence of ulcerative colitis (UC) is increasing annually. There are few treatments for UC patients, and some drugs have serious side effects. Sea cucumber peptide (SCP) has anti-inflammatory, antioxidant and other biological activities, and various sea cucumber species are in pharmaceutical development. However, relevant studies on the effects of SCP on UC progression are still lacking. In this study, a mouse model of acute colitis was induced by 3% dextran sulfate (DSS), and the effect of 500 mg/kg SCP on colitis was investigated. The results showed that SCP can alleviate DSS-induced colon damage and intestinal barrier damage. SCP significantly inhibited the expression of inflammatory factors and oxidative stress in UC mice. SCP reversed the intestinal microbiota dysregulation induced by DSS, inhibited the growth of Sutterella, Prevotella_9 and Escherichia-Shigella harmful bacteria, and increased the abundance of Lachnospiraceae_NK4A136_group. At the same time, SCP treatment significantly inhibited the LPS-induced polarization of M1 macrophages, which may be mediated by two monopeptides, IPGAPGVP and TGPIGPPGSP, via FPR2. In conclusion, SCP can protect against colitis by modulating the intestinal microbiota composition and the intestinal barrier and inhibiting the polarization of M1 macrophages.
Collapse
Affiliation(s)
- Song Yu
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| | - Haixiang Guo
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| | - Zhonghao Ji
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
- Department of Basic Medicine, Changzhi Medical College, Changzhi 046000, China
| | - Yi Zheng
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| | - Bingbing Wang
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| | - Qingqing Chen
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| | - Hongyu Tang
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| | - Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| |
Collapse
|
21
|
Xue L, Jin X, Ji T, Li R, Zhuge X, Xu F, Quan Z, Tong H, Yu W. Luteolin ameliorates DSS-induced colitis in mice via suppressing macrophage activation and chemotaxis. Int Immunopharmacol 2023; 124:110996. [PMID: 37776768 DOI: 10.1016/j.intimp.2023.110996] [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: 06/23/2023] [Revised: 09/03/2023] [Accepted: 09/24/2023] [Indexed: 10/02/2023]
Abstract
OBJECTIVES Luteolin, known for its multifaceted therapeutic properties against inflammatory diseases, holds potential for addressing the unmet need for effective treatments in ulcerative colitis (UC), a prevalent subtype of inflammatory bowel disease (IBD). This study aimed to comprehensively assess luteolin's therapeutic efficacy in a dextran sulfate sodium (DSS)-induced colitis mouse model, shedding light on its anti-UC mechanisms. METHODS Our investigation encompassed in vivo assessments of luteolin's therapeutic potential against DSS-induced colitis through rigorous histopathological examination and biochemical analyses. Furthermore, we scrutinized luteolin's anti-inflammatory prowess in vitro using lipopolysaccharide (LPS)-stimulated RAW264.7 cells and primary peritoneal macrophages. Additionally, we quantitatively evaluated the impact of luteolin on C-C motif chemokine ligand 2 (CCL2)-induced macrophage migration employing Transwell and Zigmond chambers. Furthermore, cellular thermal shift assay (CETSA), drug affinity responsive target stability (DARTS) assay, and molecular docking were employed to identify potential therapeutic targets of luteolin and investigate their binding sites and interaction patterns. RESULTS Luteolin demonstrated therapeutic potential against DSS-induced colitis by ameliorating colitis symptoms, restoring intestinal barrier integrity, and inhibiting proinflammatory cytokine production in the colonic tissues. Moreover, luteolin demonstrated robust anti-inflammatory activity in vitro, in lipopolysaccharide (LPS)-stimulated RAW264.7 cells and primary peritoneal macrophages. Notably, luteolin suppressed the phosphorylation of IKKα/β, IκBα, and p65, along with preventing IκBα degradation in LPS-treated RAW264.7 cells and peritoneal macrophages. Furthermore, luteolin impaired the migratory behavior of RAW264.7 cells and peritoneal macrophages, as evidenced by reduced migration distance and velocity of luteolin-treated macrophages. Mechanistically, luteolin was found to antagonize IKKα/β, subsequently inhibiting IKKα/β phosphorylation and the activation of NF-κB signaling. CONCLUSION Luteolin emerges as a promising lead compound for the clinical therapy of colitis by virtue of its ability to ameliorate DSS-induced colitis, antagonize IKKα/β, suppress NF-κB signaling, and impede macrophage activation and migration.
Collapse
Affiliation(s)
- Liwei Xue
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, Zhejiang Province, PR China
| | - Xiaosheng Jin
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, Zhejiang Province, PR China
| | - Tingting Ji
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, Zhejiang Province, PR China
| | - Rongzhou Li
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, Zhejiang Province, PR China
| | - Xiaoju Zhuge
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, Zhejiang Province, PR China
| | - Fang Xu
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, Zhejiang Province, PR China
| | - Zijiao Quan
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, Zhejiang Province, PR China
| | - Haibin Tong
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, Zhejiang Province, PR China.
| | - Weilai Yu
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, Zhejiang Province, PR China.
| |
Collapse
|
22
|
Sienkiewicz M, Sroka K, Binienda A, Jurk D, Fichna J. A new face of old cells: An overview about the role of senescence and telomeres in inflammatory bowel diseases. Ageing Res Rev 2023; 91:102083. [PMID: 37802318 DOI: 10.1016/j.arr.2023.102083] [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: 08/21/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023]
Abstract
Cellular senescence is a pivotal factor contributing to aging and the pathophysiology of age-related diseases. Despite the presence of inflammation and abnormal immune system function in both inflammatory bowel diseases (IBD) and senescence, the relationship between the two remains largely unexplored. Therefore, our study aimed to investigate the intricate connection between cellular senescence, telomeres, and IBD. The review highlights the presence of senescence markers, particularly p16 and p21, in IBD patients, suggesting their potential association with disease progression and mucosal inflammation. We emphasize the critical role of macrophages in eliminating senescent cells and how disturbance in effective clearance may contribute to persistent senescence and inflammation in IBD. Additionally, we shed light on the involvement of telomeres in IBD, as their dysfunction impairs enterocyte function and disrupts colonic barrier integrity, potentially exacerbating the pathogenesis of the disease. Targeting senescence and telomere dysfunctions holds promise for the development of innovative therapeutic approaches to mitigate intestinal inflammation and alleviate symptoms in IBD patients. By unraveling the precise role of senescence in IBD, we can pave the way for the discovery of novel therapeutic interventions that effectively address the underlying mechanisms of intestinal inflammation, offering hope for improved management and treatment of IBD patients.
Collapse
Affiliation(s)
- Michał Sienkiewicz
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Kamila Sroka
- Department of Family Medicine and Public Health, University of Opole, Opole, Poland
| | - Agata Binienda
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Diana Jurk
- Robert and Arlene Kogod Center On Aging, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
23
|
Yang J, Guan X, He S, Ge L, Gao Q, Wu X. FTY720 attenuates acute colitis via colonic T cells reduction and inhibition of M1 macrophages polarization independent of CCR2-mediated monocytes input. Int Immunopharmacol 2023; 123:110731. [PMID: 37541109 DOI: 10.1016/j.intimp.2023.110731] [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: 05/20/2023] [Revised: 07/11/2023] [Accepted: 07/27/2023] [Indexed: 08/06/2023]
Abstract
Ulcerative colitis (UC) is a complex multifactorial disease, of which the exact etiology is not fully understood. The inappropriate aggressive inflammatory response is closely related to the disease progression of UC. FTY720 is a sphingosine-1-phosphate receptor agonist and acts as a key immunomodulator in inflammation. This study aims to investigate the protective influence of FTY720 on inflammation in the DSS-induced colitis model. In the present study, the C57BL/6 mice and the CCR2-/- mice were exposed to 5% Dextran Sodium Sulfate (DSS) drinking water for 6 days followed by an injection of FTY720 (1 mg/kg/d) or vehicle (PBS) 6 times starting on the next day. The body weight, stool consistency, and occult blood were assessed daily. The inflammatory cytokines level in colon tissues and serum were assessed. Leukocyte subsets of bone marrow (BM), spleen, and colon were analyzed by flow cytometry. Our results demonstrated that FTY720 ameliorated the aberrant immune responses by trapping T cells and inhibiting the polarization of M1 macrophages in colitis mice. The effect of FTY720 on the increased number of colonic macrophages did not dependent on CCR2-mediated monocyte influx, despite most monocytes being reduced after DSS administration in the inflamed colon of CCR2-/- mice. Rather, depletion of CCR2 did not impact the protective influence of FTY720 on colonic injury in acute colitis. All these findings unravel a beneficial function of FTY720 in the inflammatory response to DSS-induced acute colitis, provided further insights into monocyte migration and might provide potential opportunities for UC therapeutic intervention.
Collapse
Affiliation(s)
- Jing Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Nankai University Affiliated Nankai Hospital, Tianjin, China.
| | - Xin Guan
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Nankai University Affiliated Nankai Hospital, Tianjin, China
| | - Simeng He
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lixiu Ge
- Department of Clinical Laboratory, Nankai University Affiliated Nankai Hospital, Tianjin, China
| | - Qiaoying Gao
- Department of Clinical Laboratory, Nankai University Affiliated Nankai Hospital, Tianjin, China
| | - Xiaoyang Wu
- Department of Anesthesiology and Critical Care Medicine, Nankai University Affiliated Nankai Hospital, Tianjin, China
| |
Collapse
|
24
|
Li D, Gu W, Xu H, Zhang Z, Zhao C, He C, Zhu X, Li Y. Inflammation in the peripheral blood system of Crohn's Disease. Clin Exp Med 2023; 23:2805-2812. [PMID: 36842094 DOI: 10.1007/s10238-023-01030-3] [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: 10/19/2022] [Accepted: 02/14/2023] [Indexed: 02/27/2023]
Abstract
Crohn's disease (CD) is an inflammatory bowel disease that is characterized by chronic inflammation of digestive system and has a nickname "green cancer" because of its sustained alternation of periods of flares and remissions. Here, we investigated the inflammation changes in peripheral blood system of CD patients, which are less reported in China. Peripheral blood samples of 167 CD patients and 30 healthy people, as well as their clinical information, were collected at the Second Affiliated Hospital of Soochow University. Flow cytometry was performed to analyze the ratio of CD4 T cells to CD8 T cells. Cytometric Bead Array kit was used to detect the cytokines in peripheral blood in CD patients. Moreover, the expression of inflammasomes was also detected by RT-PCR. The percentage and cell number of lymphocytes in CD patients' peripheral blood system decreased significantly, while monocytes increased remarkably. Interestingly, there was an inversion of the CD4 T cells/CD8 T cells ratio in peripheral blood of CD patients. The levels of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) increased significantly in CD patients' peripheral blood, and lipopolysaccharide (LPS) stimulation aggravate inflammatory response. In addition, the expression of nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 1 (NLRP1) and NLRP3 in peripheral blood mononuclear cells (PBMC) of CD patients increased significantly after LPS stimulation. The inflammation in peripheral blood of CD patients had significant changes, including PBMC, cytokines and inflammasomes. These results are helpful to get a deeper understanding of CD and improve the efficiency of diagnosis and treatment in China.
Collapse
Affiliation(s)
- Dan Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, People's Republic of China
| | - Wenyong Gu
- Department of Ultrasound, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, People's Republic of China
| | - Han Xu
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, People's Republic of China
| | - Zhiru Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, People's Republic of China
| | - Chenhao Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, People's Republic of China
| | - Chunyan He
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, People's Republic of China
- Institute of Laboratory Medicine, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004, Jiangsu, People's Republic of China
| | - Xueming Zhu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, People's Republic of China.
- Institute of Laboratory Medicine, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004, Jiangsu, People's Republic of China.
| | - Yang Li
- Institute of Laboratory Medicine, The Second Affiliated Hospital of Soochow University, 1055 San-Xiang Road, Suzhou, 215004, Jiangsu, People's Republic of China.
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, People's Republic of China.
| |
Collapse
|
25
|
Ding Z, Ge W, Xu X, Xu X, Wang S, Zhang J. PER2/P65-driven glycogen synthase 1 transcription in macrophages modulates gut inflammation and pathogenesis of rectal prolapse. J Biol Chem 2023; 299:105219. [PMID: 37660913 PMCID: PMC10534228 DOI: 10.1016/j.jbc.2023.105219] [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: 04/25/2023] [Revised: 08/12/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023] Open
Abstract
Rectal prolapse in serious inflammatory bowel disease is caused by abnormal reactions of the intestinal mucosal immune system. The circadian clock has been implicated in immune defense and inflammatory responses, but the mechanisms by which it regulates gut inflammation remain unclear. In this study, we investigate the role of the rhythmic gene Period2 (Per2) in triggering inflammation in the rectum and its contribution to the pathogenesis of rectal prolapse. We report that Per2 deficiency in mice increased susceptibility to intestinal inflammation and resulted in spontaneous rectal prolapse. We further demonstrated that PER2 was essential for the transcription of glycogen synthase 1 by interacting with the NF-κB p65. We show that the inhibition of Per2 reduced the levels of glycogen synthase 1 and glycogen synthesis in macrophages, impairing the capacity of pathogen clearance and disrupting the composition of gut microbes. Taken together, our findings identify a novel role for Per2 in regulating the capacity of pathogen clearance in macrophages and gut inflammation and suggest a potential animal model that more closely resembles human rectal prolapse.
Collapse
Affiliation(s)
- Zhao Ding
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Wenhao Ge
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Xiaodong Xu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Xi Xu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Shiming Wang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Jianfa Zhang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China.
| |
Collapse
|
26
|
Bartels JL, Fernandez SR, Arnold JS, Parker CC, Tekin V, O'Malley G, Ralph DA, Lapi SE. In vivo Assessment of the Impact of Molecular Weight on Constructs of 68Ga-DOTA-Manocept in a Syngeneic Mouse Tumor Model. Mol Imaging Biol 2023; 25:867-874. [PMID: 36882605 PMCID: PMC10598080 DOI: 10.1007/s11307-023-01809-6] [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: 09/09/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023]
Abstract
PURPOSE Manocept™ constructs are mannosylated amine dextrans (MADs) that bind with high affinity to the mannose receptor, CD206. Tumor-associated macrophages (TAMs) are the most numerous immune cells in the tumor microenvironment and a recognized target for tumor imaging and cancer immunotherapies. Most TAMs express CD206, suggesting utility of MADs to deliver imaging moieties or therapeutics to TAMs. The liver Kupffer cells also express CD206, making them an off-target localization site when targeting CD206 on TAMs. We evaluated TAM targeting strategies using two novel MADs differing in molecular weight in a syngeneic mouse tumor model to determine how varying MAD molecular weights would impact tumor localization. Increased mass dose of the non-labeled construct or a higher molecular weight (HMW) construct were also used to block liver localization and enhance tumor to liver ratios. PROCEDURES Two MADs, 8.7 kDa and 22.6 kDa modified with DOTA chelators, were synthesized and radiolabeled with 68Ga. A HMW MAD (300 kDa) was also synthesized as a competitive blocking agent for Kupffer cell localization. Balb/c mice, with and without CT26 tumors, underwent dynamic PET imaging for 90 min followed by biodistribution analyses in selected tissues. RESULTS The new constructs were readily synthesized and labeled with 68Ga with ≥ 95% radiochemical purity in 15 min at 65 °C. When injected at doses of 0.57 nmol, the 8.7 kDa MAD provided 7-fold higher 68Ga tumor uptake compared to the 22.6 kDa MAD (2.87 ± 0.73%ID/g vs. 0.41 ± 0.02%ID/g). Studies with increased mass of unlabeled competitors showed reduced liver localization of the [68Ga]MAD-8.7 to varying degrees without significant reductions in tumor localization, resulting in enhanced tumor to liver signal ratios. CONCLUSION Novel [68Ga]Manocept constructs were synthesized and studied in in vivo applications, showing that the smaller MAD localized to CT26 tumors more effectively than the larger MAD and that the unlabeled HMW construct could selectively block liver binding of [68Ga]MAD-8.7 without diminishing the localization to tumors. Promising results using the [68Ga]MAD-8.7 show a potential path to clinical applications.
Collapse
Affiliation(s)
- Jennifer L Bartels
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Solana R Fernandez
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jeffrey S Arnold
- Navidea Biopharmaceuticals, 4995 Bradenton Ave, Dublin, OH, 43017, USA
| | - Candace C Parker
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Volkan Tekin
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Grace O'Malley
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - David A Ralph
- Navidea Biopharmaceuticals, 4995 Bradenton Ave, Dublin, OH, 43017, USA
| | - Suzanne E Lapi
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
27
|
Xu YY, Zhu M, Wu J, Luo LB, Dong SJ, Zhang MG, Liu X, Wang K, Luo H, Jing WH, Wang L, Wang SC. A mannitol-modified emodin nano-drug restores the intestinal barrier function and alleviates inflammation in a mouse model of DSS-induced ulcerative colitis. Chin Med 2023; 18:98. [PMID: 37568235 PMCID: PMC10416390 DOI: 10.1186/s13020-023-00801-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 07/14/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is an inflammatory disease of the colon that is characterized by mucosal ulcers. Given its increasing prevalence worldwide, it is imperative to develop safe and effective drugs for treating UC. Emodin, a natural anthraquinone derivative present in various medicinal herbs, has demonstrated therapeutic effects against UC. However, low bioavailability due to poor water solubility limits its clinical applications. METHODS Emodin-borate nanoparticles (EmB) were synthesized to improve drug solubility, and they modified with oligomeric mannitol into microgels (EmB-MO) for targeted delivery to intestinal macrophages that express mannose receptors. UC was induced in a mouse model using dextran sulfate sodium (DSS), and different drug formulations were administered to the mice via drinking water. The levels of inflammation-related factors in the colon tissues and fecal matter were measured using enzyme-linked immunosorbent assay. Intestinal permeability was evaluated using fluorescein isothiocyanate dextran. HE staining, in vivo imaging, real-time PCR, and western blotting were performed to assess intestinal barrier dysfunction. RESULTS Both EmB and EmB-MO markedly alleviated the symptoms of UC, including body weight loss, stool inconsistency, and bloody stools and restored the levels of pro- and anti-inflammatory cytokines. However, the therapeutic effects of EmB-MO on the macroscopic and immunological indices were stronger than those of EmB and similar to those of 5-aminosalicylic acid. Furthermore, EmB-MO selectively accumulated in the inflamed colon epithelium and restored the levels of the gut barrier proteins such as ZO-1 and Occludin. CONCLUSIONS EmB-MO encapsulation significantly improved water solubility, which translated to greater therapeutic effects on the immune balance and gut barrier function in mice with DSS-induced UC. Our findings provide novel insights into developing emodin-derived drugs for the management of UC.
Collapse
Affiliation(s)
- Yin-Yue Xu
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, 710061 China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening and Analysis, Xi’an, 710061 China
| | - Min Zhu
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, 710061 China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening and Analysis, Xi’an, 710061 China
| | - Jiang Wu
- College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100 Shaanxi China
| | - Long-Biao Luo
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, 710061 China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Si-jing Dong
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, 710061 China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening and Analysis, Xi’an, 710061 China
| | - Meng-Gai Zhang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, 710061 China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening and Analysis, Xi’an, 710061 China
| | - Xue Liu
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, 710061 China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening and Analysis, Xi’an, 710061 China
| | - Ke Wang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, 710061 China
| | - Hua Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Wang-Hui Jing
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, 710061 China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening and Analysis, Xi’an, 710061 China
| | - Lin Wang
- College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100 Shaanxi China
- State Key Laboratory of Molecular Engineering of Polymers (Fudan University), Shanghai, 200438 China
| | - Si-Cen Wang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, 710061 China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening and Analysis, Xi’an, 710061 China
| |
Collapse
|
28
|
Zhang Y, Ma R, You C, Leng X, Wang D, Deng S, He B, Guo Z, Guan Z, Lei H, Yu J, Zhou Q, Xing J, Dong Y. Hyaluronic acid modified oral drug delivery system with mucoadhesiveness and macrophage-targeting for colitis treatment. Carbohydr Polym 2023; 313:120884. [PMID: 37182972 DOI: 10.1016/j.carbpol.2023.120884] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/26/2023] [Accepted: 04/02/2023] [Indexed: 05/16/2023]
Abstract
Based on the biocompatibility and macrophage targeting of natural polysaccharides, combined with the physiological and pathological characteristics of the gastrointestinal tract and colonic mucosa of ulcerative colitis (UC), we prepare dexamethasone (Dex)-loaded oral colon-targeted nano-in-micro drug delivery systems coated with multilayers of chitosan (CS), hyaluronic acid (HA), and finally Eudragit S100 (ECHCD MPs) using a layer-by-layer coating technique for UC treatment through regulating the M1/M2 polarization of intestinal macrophages. HA/CS/Dex nanoparticles (HCD NPs) are ingested by macrophages via CD44 receptor-mediated endocytosis to regulate M1-to-M2 macrophage polarization and exert anti-inflammatory effects. Moreover, ECHCD MPs show better colon-targeting properties than Dex-loaded chitosan nanoparticles (CD NPs) and HCD NPs which is demonstrated by stronger mucoadhesion to inflamed colon tissues. After oral administration, ECHCD MPs exert significant anti-UC effects. Therefore, ECHCD MPs are proven to be as promising oral colon-targeting drug delivery systems for Dex and have potential application in UC treatment.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ruirui Ma
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Cuiyu You
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xue Leng
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Danyang Wang
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shujing Deng
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Binyang He
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ziyang Guo
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zelin Guan
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hengyu Lei
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jie Yu
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qinyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jianfeng Xing
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Yalin Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
29
|
Zhang Y, Lei H, Wang P, Zhou Q, Yu J, Leng X, Ma R, Wang D, Dong K, Xing J, Dong Y. Restoration of dysregulated intestinal barrier and inflammatory regulation through synergistically ameliorating hypoxia and scavenging reactive oxygen species using ceria nanozymes in ulcerative colitis. Biomater Res 2023; 27:75. [PMID: 37507801 PMCID: PMC10375752 DOI: 10.1186/s40824-023-00412-8] [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: 01/10/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Reactive oxygen species (ROS) overproduction and excessive hypoxia play pivotal roles in the initiation and progression of ulcerative colitis (UC). Synergistic ROS scavenging and generating O2 could be a promising strategy for UC treatment. METHODS Ceria nanozymes (PEG-CNPs) are fabricated using a modified reverse micelle method. We investigate hypoxia attenuating and ROS scavenging of PEG-CNPs in intestinal epithelial cells and RAW 264.7 macrophages and their effects on pro-inflammatory macrophages activation. Subsequently, we investigate the biodistribution, pharmacokinetic properties and long-term toxicity of PEG-CNPs in mice. PEG-CNPs are administered intravenously to mice with 2,4,6-trinitrobenzenesulfonic acid-induced colitis to test their colonic tissue targeting and assess their anti-inflammatory activity and mucosal healing properties in UC. RESULTS PEG-CNPs exhibit multi-enzymatic activity that can scavenge ROS and generate O2, promote intestinal epithelial cell healing and inhibit pro-inflammatory macrophage activation, and have good biocompatibility. After intravenous administration of PEG-CNPs to colitis mice, they can enrich at the site of colonic inflammation, and reduce hypoxia-induced factor-1α expression in intestinal epithelial cells by scavenging ROS to generate O2, thus further promoting disrupted intestinal mucosal barrier restoration. Meanwhile, PEG-CNPs can effectively scavenge ROS in impaired colon tissues and relieve colonic macrophage hypoxia to suppress the pro-inflammatory macrophages activation, thereby preventing UC occurrence and development. CONCLUSION This study has provided a paradigm to utilize metallic nanozymes, and suggests that further materials engineering investigations could yield a facile method based on the pathological characteristics of UC for clinically managing UC.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Hengyu Lei
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Pengchong Wang
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Qinyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jie Yu
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xue Leng
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ruirui Ma
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Danyang Wang
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Kai Dong
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Jianfeng Xing
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Yalin Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
30
|
Zhang X, Wang P, Yang Y, Gao X, Tang Y, Luo Y, Deng X, Yu Y, Zhang L, Zhang Y. Monocyte/macrophage mediates the impact of depression on Crohn's disease. Scand J Gastroenterol 2023; 58:1434-1444. [PMID: 37635462 DOI: 10.1080/00365521.2023.2245938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/28/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Depression increases the risk of Crohn's disease (CD) and worsens its prognosis. Monocytes/macrophages, immune modulate cells, play vital roles in both depression and CD. OBJECTIVES We investigated whether monocyte/macrophage could mediate the impact of depression on CD through induction of CD4 + T lymphocyte differentiation and epithelial barrier dysfunction, in addition to the alteration of their own phagocytic ability and cytokines production. METHODS Circulating monocytes and intestinal macrophages were isolated from eligible CD patients, divided into depressed and non-depressed groups. Phagocytosis was determined using flow cytometry while in vitro cytokine production was quantified using Luminex assay and qPCR. CD4 + T cells were cocultured with monocytes, then Type 1 Helper T Lymphocytes Th1/Type 2 Helper T Lymphocytes (Th2) /Type 17 Helper T Lymphocytes (Th17)/Treg subsets were analyzed using flow cytometry and qPCR. Caco-2 monolayers simulating epithelial barrier were cocultured with macrophages, and integrity and proliferation were evaluated. Tight junction protein expression was detected using immunofluorescence and western blot. RESULTS Decreased monocyte/macrophage phagocytosis and enhanced production of pro-inflammatory cytokines including Tumor necrosis factor-α (TNF-α), Interleukin-6 (IL-6) and Interleukin-1β (IL-1β) were revealed in the depressed versus non-depressed CD groups. Higher proportions of Th1 and Th17 cells with a lower proportion of Treg cell were observed after cocultured with monocytes from the depressed versus non-depressed CD patients. So were the expressions of their corresponding transcription factors T-bet, Retinoic Acid Related Orphan Nuclear Receptor gamma T (RORγt) and Forkhead box protein P3 (FoxP3). Caco-2 cells cocultured with macrophages from depressed CD displayed lower Transepithelial electric resistance (TEER), reduced proliferation activity, and decreased tight junction protein expressions compared with their counterpart cocultured with macrophages from non-depressed CD. CONCLUSIONS Monocyte/macrophage may underlie the impact of depression upon CD via decreased phagocytosis, increased pro-inflammatory cytokine production, inducing CD4 + T cell differentiation toward Th1/Th17 cells rather than Treg cell, and impairing macrophage-enhanced epithelial barrier.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Pengbo Wang
- School of Professional Studies, Columbia University, New York, NY, USA
| | - Yi Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xin Gao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Yu Tang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Ying Luo
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiangbing Deng
- Department of Gastrointestinal Surgery, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Yongyang Yu
- Department of Gastrointestinal Surgery, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Lanlan Zhang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, Chengdu, People's Republic of China
| | - Yan Zhang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
31
|
Zhao D, Qin D, Yin L, Yang Q. Integrated Bioinformatics Analysis and Experimental Verification of Immune Cell Infiltration and the Related Core Genes in Ulcerative Colitis. Pharmgenomics Pers Med 2023; 16:629-643. [PMID: 37383675 PMCID: PMC10296601 DOI: 10.2147/pgpm.s406644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/26/2023] [Indexed: 06/30/2023] Open
Abstract
Background Ulcerative colitis is a recurrent autoimmune disease. At present, the pathogenesis of UC is not completely clear. Hence, the etiology and underlying molecular mechanism need to be further investigated. Methods Three sets of microarray datasets were included from the Gene Expression Omnibus database. The differentially expressed genes in two sets of datasets were analyzed using the R software, and the core genes of UC were screened using machine learning. The sensitivity and specificity of the core genes were evaluated with the receiver operating characteristic curve in another microarray dataset. Subsequently, the CIBERSORT tool was used to analyze the relationship between UC and its core genes and immune cell infiltration. To verify the relationship between UC and core genes and the relationship between core genes and immune cell infiltration in vivo. Results A total of 36 DEGs were identified. AQP8, HMGCS2, and VNN1 were determined to be the core genes of UC. These genes had high sensitivity and specificity in receiver operating characteristic curve analysis. According to the analysis of immune cell infiltration, neutrophils, monocytes, and macrophages were positively correlated with UC. AQP8, HMGCS2, and VNN1 were also correlated with immune cell infiltration to varying degrees. In vivo experiments verified that the expressions of neutrophils, monocytes, and macrophages increased in the UC colon. Furthermore, the expressions of AQP8 and HMGCS2 decreased, whereas that of VNN1 increased. Azathioprine treatment improved all the indicators to different degrees. Conclusion AQP8, HMGCS2, and VNN1 are the core genes of UC and exhibit different degrees of correlation with immune cells. These genes are expected to become new therapeutic targets for UC. Moreover, the occurrence and development of UC are influenced by immune cell infiltration.
Collapse
Affiliation(s)
- Danya Zhao
- The First School of Clinical Medicine of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Danping Qin
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Liming Yin
- Institute of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Qiang Yang
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| |
Collapse
|
32
|
Du J, Zhang J, Wang L, Wang X, Zhao Y, Lu J, Fan T, Niu M, Zhang J, Cheng F, Li J, Zhu Q, Zhang D, Pei H, Li G, Liang X, Huang H, Cao X, Liu X, Shao W, Sheng J. Selective oxidative protection leads to tissue topological changes orchestrated by macrophage during ulcerative colitis. Nat Commun 2023; 14:3675. [PMID: 37344477 PMCID: PMC10284839 DOI: 10.1038/s41467-023-39173-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 06/01/2023] [Indexed: 06/23/2023] Open
Abstract
Ulcerative colitis is a chronic inflammatory bowel disorder with cellular heterogeneity. To understand the composition and spatial changes of the ulcerative colitis ecosystem, here we use imaging mass cytometry and single-cell RNA sequencing to depict the single-cell landscape of the human colon ecosystem. We find tissue topological changes featured with macrophage disappearance reaction in the ulcerative colitis region, occurring only for tissue-resident macrophages. Reactive oxygen species levels are higher in the ulcerative colitis region, but reactive oxygen species scavenging enzyme SOD2 is barely detected in resident macrophages, resulting in distinct reactive oxygen species vulnerability for inflammatory macrophages and resident macrophages. Inflammatory macrophages replace resident macrophages and cause a spatial shift of TNF production during ulcerative colitis via a cytokine production network formed with T and B cells. Our study suggests components of a mechanism for the observed macrophage disappearance reaction of resident macrophages, providing mechanistic hints for macrophage disappearance reaction in other inflammation or infection situations.
Collapse
Affiliation(s)
- Juan Du
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China.
| | - Junlei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang University Cancer Centre, Zhejiang University, Hangzhou, 310002, China
| | - Lin Wang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang University Cancer Centre, Zhejiang University, Hangzhou, 310002, China
| | - Xun Wang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang University Cancer Centre, Zhejiang University, Hangzhou, 310002, China
| | - Yaxing Zhao
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang University Cancer Centre, Zhejiang University, Hangzhou, 310002, China
| | - Jiaoying Lu
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Tingmin Fan
- Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Central Laboratory, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310002, China
| | - Meng Niu
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Jie Zhang
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Fei Cheng
- Pathology Department, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Jun Li
- Pathology Department, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Qi Zhu
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, No. 29 JiangJun Road, Jiang Ning District, Nanjing, Jiangsu, 211106, China
| | - Daoqiang Zhang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, No. 29 JiangJun Road, Jiang Ning District, Nanjing, Jiangsu, 211106, China
| | - Hao Pei
- MobiDrop (Zhejiang), No. 455 Heshun Road, Tongxiang, Zhejiang, 314500, China
| | - Guang Li
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Chaoyang District, Beijing, 100024, China
| | - Xingguang Liang
- Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Central Laboratory, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310002, China
| | - He Huang
- Frontiers Science Center for Synthetic Biology, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Xiaocang Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Department of Hepato-Gastroenterology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300000, China.
| | - Xinjuan Liu
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Chaoyang District, Beijing, 100024, China.
| | - Wei Shao
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, No. 29 JiangJun Road, Jiang Ning District, Nanjing, Jiangsu, 211106, China.
| | - Jianpeng Sheng
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China.
- Zhejiang University Cancer Centre, Zhejiang University, Hangzhou, 310002, China.
| |
Collapse
|
33
|
Zhou L, Yan Z, Yang W, Buckley JA, Al Diffalha S, Benveniste EN, Qin H. Socs3 expression in myeloid cells modulates the pathogenesis of dextran sulfate sodium (DSS)-induced colitis. Front Immunol 2023; 14:1163987. [PMID: 37283760 PMCID: PMC10239850 DOI: 10.3389/fimmu.2023.1163987] [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: 02/11/2023] [Accepted: 05/05/2023] [Indexed: 06/08/2023] Open
Abstract
Introduction Myeloid cells play a critical role in the pathogenesis of Inflammatory Bowel Diseases (IBDs), including Ulcerative Colitis (UC) and Crohn's Disease (CD). Dysregulation of the JAK/STAT pathway is associated with many pathological conditions, including IBD. Suppressors Of Cytokine Signaling (SOCS) are a family of proteins that negatively regulate the JAK/STAT pathway. Our previous studies identified that mice lacking Socs3 in myeloid cells developed a hyper-activated phenotype of macrophages and neutrophils in a pre-clinical model of Multiple Sclerosis. Methods To better understand the function of myeloid cell Socs3 in the pathogenesis of colitis, mice with Socs3 deletion in myeloid cells (Socs3 ΔLysM) were utilized in a DSS-induced colitis model. Results Our results indicate that Socs3 deficiency in myeloid cells leads to more severe colitis induced by DSS, which correlates with increased infiltration of monocytes and neutrophils in the colon and increased numbers of monocytes and neutrophils in the spleen. Furthermore, our results demonstrate that the expression of genes related to the pathogenesis and diagnosis of colitis such as Il1β, Lcn2, S100a8 and S100a9 were specifically enhanced in Socs3-deficient neutrophils localized to the colon and spleen. Conversely, there were no observable differences in gene expression in Ly6C+ monocytes. Depletion of neutrophils using a neutralizing antibody to Ly6G significantly improved the disease severity of DSS-induced colitis in Socs3-deficient mice. Discussion Thus, our results suggest that deficiency of Socs3 in myeloid cells exacerbates DSS-induced colitis and that Socs3 prevents overt activation of the immune system in IBD. This study may provide novel therapeutic strategies to IBD patients with hyperactivated neutrophils.
Collapse
Affiliation(s)
- Lianna Zhou
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Zhaoqi Yan
- Gladstone Institute of Neurological Disease, San Francisco, CA, United States
| | - Wei Yang
- Division of Gastroenterology and Hepatology, Weill Cornell College of Medicine, New York, NY, United States
| | - Jessica A. Buckley
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sameer Al Diffalha
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Etty N. Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
34
|
Feng B, Xu L, Song S, Liu H, Li Y, Hu S, Shu Q, Liu J, Liu Z, Yu H, Yang P. ER stress modulates the immune regulatory ability in gut M2 cells of patients with ulcerative colitis. iScience 2023; 26:106498. [PMID: 37091242 PMCID: PMC10113856 DOI: 10.1016/j.isci.2023.106498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/28/2023] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
This study aims to characterize the impaired immune regulatory function of Mφ obtained from UC patient colon lavage fluid (CLF). Mφs were the largest proportion (21.3 4.0%) of the CLF-derived cellular components. Less abundant and weaker immune suppressive function were observed in M2 Mφs (M2 cells) of the ulcerative colitis (UC) group. High levels of endoplasmic reticulum (ER) stress associated molecules were detected in UC M2 cells. The spliced X box binding protein-1 (XBP1) gene was negatively correlated with programmed death ligand-1 (PD-L1) in UC M2 cells. XBP1 promoted the expression of ring-finger protein 20 (Rnf20) in M2 cells. Rnf20 reduced PD-L1 abundance in UC M2 cells and impaired the immune suppressive ability. Inhibition of Rnf20 restored the immune regulating capacity of M2 cells and suppressed experimental colitis.
Collapse
Affiliation(s)
- Baisui Feng
- Department of Gastroenterology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lingzhi Xu
- Department of Immunology, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Shuo Song
- Guangdong Provincial Regional Disease Key Laboratory, Shenzhen, China
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Division at Shenzhen University, Shenzhen, China
| | - Huazhen Liu
- Guangdong Provincial Regional Disease Key Laboratory, Shenzhen, China
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Division at Shenzhen University, Shenzhen, China
| | - Yan Li
- Department of Gastroenterology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Suqin Hu
- Guangdong Provincial Regional Disease Key Laboratory, Shenzhen, China
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Division at Shenzhen University, Shenzhen, China
| | - Qing Shu
- Department of Gastroenterology, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Jiangqi Liu
- Department of Allergy, Longgang ENT Hospital, Shenzhen, China
| | - Zhiqiang Liu
- Department of Allergy, Longgang ENT Hospital, Shenzhen, China
| | - Haiqiong Yu
- Department of Pulmonary and Critical Care Medicine, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen518033, China
- Corresponding author
| | - Pingchang Yang
- Guangdong Provincial Regional Disease Key Laboratory, Shenzhen, China
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Division at Shenzhen University, Shenzhen, China
- Corresponding author
| |
Collapse
|
35
|
Xiong K, Deng J, Yue T, Hu W, Zeng X, Yang T, Xiao T. Berberine promotes M2 macrophage polarisation through the IL-4-STAT6 signalling pathway in ulcerative colitis treatment. Heliyon 2023; 9:e14176. [PMID: 36923882 PMCID: PMC10009548 DOI: 10.1016/j.heliyon.2023.e14176] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023] Open
Abstract
Aim This study focusses on the anti-inflammatory and immune-modulatory roles of berberine (BBR) in ulcerative colitis (UC) treatment. Additionally, the underlying mechanisms of BBR were systematically explored. Methods A 3% (w/v) dextran sodium sulphate (DSS) solution was used for establishing the mice UC model. M2 macrophage polarisation was induced in RAW 264.7 cells using interleukin 4 (IL-4), whereas M1 macrophage polarisation was induced using lipopolysaccharide. Colon length, colon mucosa damage index (CMDI), and haematoxylin-eosin (HE) staining were used to evaluate colon damage induced by DSS. M1/M2 macrophages in the colon tissue were identified using immunofluorescence (IF) staining with CD86+ or CD163+. M1/M2 macrophages in the abdomen were examined using flow cytometry. An enzyme-linked immunosorbent assay was conducted to identify M1/M2 macrophage supernatant biomarkers in RAW 264.7 cells. Western blotting, immunohistochemical staining, and real-time PCR were performed to investigate the potential mechanisms of BBR for treating UC in vivo and in vitro. Results BBR was found to prolong colon length, ameliorate CMDI and alleviate the colon's pathological changes in UC mice. In DSS-induced UC mice, M1 macrophages predominated. BBR promoted M2 macrophages and suppressed M1 macrophages in the colon and abdomen of DSS-induced UC mice. Additionally, BBR significantly decreased M1-specific markers (IFN-γ and IL-1β) while increasing M2-specific markers (IL-10 and TGF-β) in the supernatants of RAW 264.7 cells. BBR upregulated the mRNA expression of IL-4, STAT6, and Chil3 while downregulating TNF-α, IFN-γ, and NOS2 expression in vivo. Moreover, BBR activated the downstream targets of the IL-4-STAT6 signalling pathway and enhanced the phosphorylation of STAT6 in vivo and in vitro to polarise M2 macrophage. Conclusion In UC mice, BBR suppressed M1 macrophages while promoting M2 macrophages. M1 macrophage suppression and M2 macrophage activation were strongly correlated with the anti-inflammatory and immune-modulating activities of BBR. BBR induced the polarisation of M2 macrophages by activating the IL-4-STAT6 signalling pathway, which contributed to its therapeutic efficacy against UC.
Collapse
Affiliation(s)
- Kai Xiong
- Colorectal and Anal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No 71 Baoshan North Road, Guiyang, 550001, China
| | - Jia Deng
- Colorectal and Anal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No 71 Baoshan North Road, Guiyang, 550001, China
| | - Tinghui Yue
- Colorectal and Anal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No 71 Baoshan North Road, Guiyang, 550001, China
| | - Wenting Hu
- Colorectal and Anal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No 71 Baoshan North Road, Guiyang, 550001, China
| | - Xinglin Zeng
- Colorectal and Anal Surgery, Chengdu Anorectal Hospital, Chengdu, 610075, China
| | - Tao Yang
- Colorectal and Anal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No 71 Baoshan North Road, Guiyang, 550001, China
| | - Tianbao Xiao
- Colorectal and Anal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No 71 Baoshan North Road, Guiyang, 550001, China
| |
Collapse
|
36
|
Yousaf H, Khan MIU, Ali I, Munir MU, Lee KY. Emerging role of macrophages in non-infectious diseases: An update. Biomed Pharmacother 2023; 161:114426. [PMID: 36822022 DOI: 10.1016/j.biopha.2023.114426] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/23/2023] Open
Abstract
In the past three decades, a huge body of evidence through various research studies conducted on animal models, has demonstrated that the macrophages are centralized of all the leukocytes involved in diseases and, particularly, their role in non-infectious diseases has been studied extensively for which they have also been referred to as the "double-edged swords". The most versatile of all immunocytes, macrophages play a key role in health and diseases. Various experimental models have demonstrated the conventional paradigms such as the M1/M2 dichotomy, which is not as obvious and presents a complex characterization of the macrophages in the disease immunology. In human diseases, this M1-M2 continuum shows a complex web of mechanisms, which are majorly divided into the pro-inflammatory roles (derived mainly by the cytokines: IL-1, IL-6, IL-12, IL-23, and tumor necrosis factor) and anti-inflammatory roles (CCl-17, CCl-22, CCL-2, transforming growth factor (TGF), and interleukin-10), which are involved in the wound healing and pathogen-suppression. The conventional division of these macrophages as M1 and M2 is derived from the opposing functions of these macrophages; where M1 is involved in the tissue damage and pro-inflammatory roles and M2 promotes cell proliferation and the resolution of inflammation. Both these pathways down-regulate each other in diseases through a plethora of enzymatic and cytokine mediators.
Collapse
Affiliation(s)
- Hassan Yousaf
- Institute of Molecular Biology and Biotechnology (IMBB), University of Lahore, Lahore, Pakistan
| | - Malik Ihsan Ullah Khan
- Institute of Molecular Biology and Biotechnology (IMBB), University of Lahore, Lahore, Pakistan.
| | - Iftikhar Ali
- Department of Life Sciences, University of Management and Technology, Lahore, Pakistan
| | - Muhammad Usman Munir
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University Sakaka, Aljouf 72388, Saudi Arabia
| | - Ka Yiu Lee
- Swedish Winter Sports Research Centre, Department of Health Sciences, Mid Sweden University, Ostersund, Sweden.
| |
Collapse
|
37
|
Ge S, Yang Y, Zuo L, Song X, Wen H, Geng Z, He Y, Xu Z, Wu H, Shen M, Ge Y, Sun X. Sotetsuflavone ameliorates Crohn's disease-like colitis by inhibiting M1 macrophage-induced intestinal barrier damage via JNK and MAPK signalling. Eur J Pharmacol 2023; 940:175464. [PMID: 36566007 DOI: 10.1016/j.ejphar.2022.175464] [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: 06/22/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Intestinal inflammation and intestinal barrier dysfunction are two important pathological changes in Crohn's disease (CD). Sotetsuflavone (SF) is a natural monomeric herbal compound with anti-inflammatory and cytoprotective effects that is mostly nontoxic. The effect of SF on CD-like spontaneous colitis was investigated in this study. METHODS Il-10-/- mice were used as a CD model and were administered different doses of SF. Lipopolysaccharide (LPS) plus IFN-γ-induced macrophages (RAW264.7) and a coculture system (RAW264.7 and organoids) were used in vitro. The protective effects of SF against CD-like colitis and macrophage differentiation and the mechanisms were evaluated. RESULTS SF treatment markedly improved spontaneous colitis in the CD model, as shown by the following evidence: reductions in the DAI, macroscopic scores (3.63 ± 1.30), colonic tissue inflammatory scores (2 ± 0.76) and proinflammatory factor levels and the attenuation of colon shortening (8 ± 0.93 cm) and weight loss (1.75 ± 1.83 g). Decreased intestinal permeability and intestinal bacterial translocation rates provided evidence of the protective effect of SF on intestinal barrier function. We also found that SF suppressed M1 macrophage-induced inflammatory responses. In the coculture system of mouse colonic organoids and RAW264.7 cells, SF significantly ameliorated M1 macrophage-induced intestinal epithelial damage. In addition, SF inhibited JNK and MAPK (p38) signalling in both Il-10-/- mice and LPS plus IFN-γ-induced macrophages (RAW264.7). CONCLUSIONS The protective effects of SF against CD-like colitis may be achieved partially by inhibiting M1 macrophage-induced intestinal barrier damage via JNK and p38 signalling. SF may have therapeutic potential for treating CD, especially considering its safety.
Collapse
Affiliation(s)
- Sitang Ge
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, China; Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yating Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Lugen Zuo
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Xue Song
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Hexin Wen
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Zhijun Geng
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yifan He
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Zilong Xu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Huatao Wu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Mengdi Shen
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Yuanyuan Ge
- Department of Colorectal Surgery, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuejun Sun
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, China.
| |
Collapse
|
38
|
Deng C, Hu Y, Conceição M, Wood MJA, Zhong H, Wang Y, Shao P, Chen J, Qiu L. Oral delivery of layer-by-layer coated exosomes for colitis therapy. J Control Release 2023; 354:635-650. [PMID: 36634710 DOI: 10.1016/j.jconrel.2023.01.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/14/2023]
Abstract
Mesenchymal stem cell-derived exosomes (MSC-Exos) have attracted much attention as a potential cell-free therapy for ulcerative colitis (UC), mainly due to their anti-inflammatory, tissue repair, and immunomodulatory properties. Although intravenous injection of MSC-Exos is able to improve UC to a certain extent, oral administration of exosomes is the preferred method to treat gastrointestinal diseases such as UC. However, exosomes contain proteins and nucleic acids that are vulnerable to degradation by the gastrointestinal environment, making oral administration difficult to implement. Layer-by-layer (LbL) self-assembly technology provides a promising strategy for the oral delivery of exosomes. Therefore, an efficient LbL-Exos self-assembly system was constructed in this study for the oral delivery of exosomes targeted to the colon to improve UC treatment. Biocompatible and biodegradable N-(2-hydroxyl) propyl-3-trimethyl ammonium chitosan chloride (HTCC) and oxidized konjac glucomannan (OKGM) polysaccharides were used as the outer layers to provide colon targeting and to protect exosomes from degradation. Similar to plain exosomes, LbL-Exos had a similar structure and features, but LbL provided controlled release of exosomes in the inflammatory colon. Compared with intravenous administration, oral administration of LbL-Exos could effectively alleviate UC using half the number of exosomes. Mechanistic studies showed that LbL-Exos were internalized by macrophages and intestinal epithelial cells to exert anti-inflammatory and tissue repair effects and therefore alleviate UC. Furthermore, the LbL-Exos system was able to improve UC via MAPK/NF-κB signaling pathway inhibition. Overall, our data show that LbL-MSC-Exos can alleviate UC after oral administration and therefore may constitute a new strategy for UC treatment in the future.
Collapse
Affiliation(s)
- Chao Deng
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Yiwei Hu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; Jiangyin Center for Disease Control and Prevention, Jiangyin 214434, China
| | | | - Matthew J A Wood
- Department of Paediatrics, University of Oxford, Oxford OX1 3QX, UK; MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Hongyao Zhong
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Yan Wang
- Yixing Hospital of Traditional Chinese Medicine, Wuxi 214200, China
| | - Ping Shao
- Yixing Hospital of Traditional Chinese Medicine, Wuxi 214200, China.
| | - Jinghua Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China.
| | - Lipeng Qiu
- Department of Paediatrics, University of Oxford, Oxford OX1 3QX, UK; School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
39
|
Yang L, Shen WW, Shao W, Zhao Q, Pang GZ, Yang Y, Tao XF, Zhang WP, Mei Q, Shen YX. MANF ameliorates DSS-induced mouse colitis via restricting Ly6C hiCX3CR1 int macrophage transformation and suppressing CHOP-BATF2 signaling pathway. Acta Pharmacol Sin 2023; 44:1175-1190. [PMID: 36635421 DOI: 10.1038/s41401-022-01045-8] [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: 08/28/2022] [Accepted: 12/19/2022] [Indexed: 01/14/2023] Open
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF), an endoplasmic reticulum stress-inducible secreting protein, has evolutionarily conserved immune-regulatory function that contributes to the negative regulation of inflammation in macrophages. In this study, we investigated the profiles of MANF in the macrophages of the patients with active inflammatory bowel disease (IBD) and the mice with experimental colitis, which was induced in both myeloid cell-specific MANF knockout mice and wild-type mice by 3% dextran sodium sulfate (DSS) for 7 days. We found that MANF expression was significantly increased in intestinal macrophages from both the mice with experimental colitis and patients with active IBD. DSS-induced colitis was exacerbated in myeloid cell-specific MANF knockout mice. Injection of recombinant human MANF (rhMANF, 10 mg·kg-1·d-1, i.v.) from D4 to D6 significantly ameliorated experimental colitis in DSS-treated mice. More importantly, MANF deficiency in myeloid cells resulted in a dramatic increase in the number of Ly6ChiCX3CRint proinflammatory macrophages in colon lamina propria of DSS-treated mice, and the proinflammatory cytokines and chemokines were upregulated as well. Meanwhile, we demonstrated that MANF attenuated Th17-mediated immunopathology by inhibiting BATF2-mediated innate immune response and downregulating CXCL9, CXCL10, CXCL11 and IL-12p40; MANF functioned as a negative regulator in inflammatory macrophages via inhibiting CHOP-BATF2 signaling pathway, thereby protecting against DSS-induced mouse colitis. These results suggest that MANF ameliorates colon injury by negatively regulating inflammatory macrophage transformation, which shed light on a potential therapeutic target for IBD.
Collapse
Affiliation(s)
- Lin Yang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Institute, Anhui Medical University, Hefei, 230032, China
| | - Wen-Wen Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Institute, Anhui Medical University, Hefei, 230032, China
| | - Wei Shao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Institute, Anhui Medical University, Hefei, 230032, China
| | - Qing Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Institute, Anhui Medical University, Hefei, 230032, China
| | - Gao-Zong Pang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Institute, Anhui Medical University, Hefei, 230032, China
| | - Yi Yang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Institute, Anhui Medical University, Hefei, 230032, China.,First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Xiao-Fang Tao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Institute, Anhui Medical University, Hefei, 230032, China
| | - Wei-Ping Zhang
- First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Qiong Mei
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Institute, Anhui Medical University, Hefei, 230032, China
| | - Yu-Xian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China. .,Biopharmaceutical Institute, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
40
|
Wang K, Mao T, Lu X, Wang M, Yun Y, Jia Z, Shi L, Jiang H, Li J, Shi R. A potential therapeutic approach for ulcerative colitis: targeted regulation of macrophage polarization through phytochemicals. Front Immunol 2023; 14:1155077. [PMID: 37197668 PMCID: PMC10183582 DOI: 10.3389/fimmu.2023.1155077] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/20/2023] [Indexed: 05/19/2023] Open
Abstract
Ulcerative colitis (UC), a type of inflammatory bowel disease characterized by recurring and incurable symptoms, causes immense suffering and economic burden for patients due to the limited treatment options available. Therefore, it is imperative to develop novel and promising strategies, as well as safe and effective drugs, for the clinical management of UC. Macrophages play a critical role as the initial line of defense in maintaining intestinal immune homeostasis, and their phenotypic transformation significantly influences the progression of UC. Scientific studies have demonstrated that directing macrophage polarization toward the M2 phenotype is an effective strategy for the prevention and treatment of UC. Phytochemicals derived from botanical sources have garnered the interest of the scientific community owing to their distinct bioactivity and nutritional value, which have been shown to confer beneficial protective effects against colonic inflammation. In this review, we explicated the influence of macrophage polarization on the development of UC and collated data on the significant potential of natural substances that can target the macrophage phenotype and elucidate the possible mechanism of action for its treatment. These findings may provide novel directions and references for the clinical management of UC.
Collapse
Affiliation(s)
- Ke Wang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tangyou Mao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyu Lu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Muyuan Wang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yifei Yun
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zeyu Jia
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Shi
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Haoxi Jiang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Junxiang Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Junxiang Li, ; Rui Shi,
| | - Rui Shi
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Junxiang Li, ; Rui Shi,
| |
Collapse
|
41
|
Xu Y, Qian W, Huang L, Wen W, Li Y, Guo F, Zhu Z, Li Z, Gong J, Yu Z, Zhou Y, Lu N, Zhu W, Guo Z. Crohn's disease-associated AIEC inhibiting intestinal epithelial cell-derived exosomal let-7b expression regulates macrophage polarization to exacerbate intestinal fibrosis. Gut Microbes 2023; 15:2193115. [PMID: 36945126 PMCID: PMC10038049 DOI: 10.1080/19490976.2023.2193115] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
The interaction between adherent-invasive Escherichia coli (AIEC) and intestinal macrophages is implicated in the pathogenesis of Crohn's disease (CD). However, its role in intestinal fibrogenesis and the underlying molecular mechanisms are poorly understood. In addition, miRNAs such as let-7b may participate in AIEC-macrophage interactions. In this study, we identified that the colonization of AIEC in the ileum was associated with enhanced intestinal fibrosis and reduced let-7b expression by enrolling a prospective cohort of CD patients undergoing ileocolectomy. Besides, AIEC-infected IL-10-/- mice presented more severe intestinal fibrosis and could be improved by exogenous let-7b. Mechanistically, intestinal macrophages were found to be the main target of let-7b. Transferring let-7b-overexpressing macrophages to AIEC-infected IL-10-/- mice significantly alleviated intestinal fibrosis. In vitro, AIEC suppressed exosomal let-7b derived from intestinal epithelial cells (IECs), instead of the direct inhibition of let-7b in macrophages, to promote macrophages to a fibrotic phenotype. Finally, TGFβR1 was identified as one target of let-7b that regulates macrophage polarization. Overall, the results of our work indicate that AIEC is associated with enhanced intestinal fibrosis in CD. AIEC could inhibit exosomal let-7b from IECs to promote intestinal macrophages to a fibrotic phenotype and then contributed to fibrogenesis. Thus, anti-AIEC or let-7b therapy may serve as novel therapeutic approaches to ameliorate intestinal fibrosis.
Collapse
Affiliation(s)
- Yihan Xu
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenwei Qian
- Department of General Surgery, Jinling Hospital, Medical School of Southeast University, Nanjing, China
| | - Liangyu Huang
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Weiwei Wen
- Department of General Surgery, Jinling Hospital, Medical School of Southeast University, Nanjing, China
| | - Yi Li
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Feilong Guo
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhenxing Zhu
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhun Li
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jianfeng Gong
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zeqian Yu
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yan Zhou
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Nan Lu
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Weiming Zhu
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhen Guo
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
42
|
He J, Liu L, Liu X, Chen H, Liu K, Huang N, Wang Y. Epoxymicheliolide prevents dextran sulfate sodium-induced colitis in mice by inhibiting TAK1-NF-κB pathway and activating Keap1-NRF2 signaling in macrophages. Int Immunopharmacol 2022; 113:109404. [PMID: 36461599 DOI: 10.1016/j.intimp.2022.109404] [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: 07/07/2022] [Revised: 10/11/2022] [Accepted: 10/28/2022] [Indexed: 11/11/2022]
Abstract
Ulcerative colitis (UC) is an unspecific colorectal inflammation associated with macrophages overactivation. Therefore, macrophage-targeted treatment has been considered a promising strategy for UC therapy. Epoxymicheliolide (EMCL) is a compound from Aucklandia lappa Decne, a TCM for treating gastrointestinal inflammatory diseases. The purpose of this study is to investigate the therapeutic effect of EMCL on DSS-induced mice colitis through the anti-inflammatory activity on macrophages and its underlying mechanisms. We found that EMCL inhibited the release of NO and PGE2 by down-regulating the expression of iNOS and COX2, while suppressed the expression of IL-1β, IL-6, and TNF-α in LPS-stimulated RAW264.7 macrophages. EMCL also inhibited NO production in LPS-activated peritoneal macrophages and TNFα-stimulated RAW264.7 cells. Moreover, EMCL blocked the phosphorylation of TAK1, IKKα/β, and IκBα, as well as IκBα degradation, thereby inhibiting the NF-κB pro-inflammatory signaling. Furthermore, EMCL decreased the intracellular ROS, by activating the NRF2 antioxidant pathway. CETSA and molecular docking showed that EMCL might form a covalent bond with Cys174 of TAK1 or Cya151 of Keap1, which may contribute to EMCL-mediated actions. Additionally, a thiol donor β-mercaptoethanol obviously abolished EMCL-mediated actions in vitro, suggesting the crucial role of the α, γ-unsaturated lactone of EMCL on its anti-inflammatory effects. Furthermore, EMCL not only ameliorated symptoms of colitis and colon barrier injury, but also decreased the levels of pro-inflammatory cytokines, MPO, NO, and MDA in DSS-challenged mice. Thus, our study demonstrated that EMCL ameliorated UC by targeting NF-κB and Nrf2 pathways, indicating it may server as a promising drug candidate for UC therapy.
Collapse
Affiliation(s)
- Jinchen He
- Department of Pathophysiology, West China College of Basic Medical Sciences & Forensic Medicine, Sichuan University, 610041 Chengdu, China
| | - Lu Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137 Chengdu, China
| | - Xiaojun Liu
- Department of Pathophysiology, West China College of Basic Medical Sciences & Forensic Medicine, Sichuan University, 610041 Chengdu, China
| | - Hongqing Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, 610072 Chengdu, China
| | - Keyun Liu
- Department of Physiology, School of Medicine, Hubei University for Nationalities, 445000 Enshi, China
| | - Ning Huang
- Department of Pathophysiology, West China College of Basic Medical Sciences & Forensic Medicine, Sichuan University, 610041 Chengdu, China.
| | - Yi Wang
- Department of Pathophysiology, West China College of Basic Medical Sciences & Forensic Medicine, Sichuan University, 610041 Chengdu, China.
| |
Collapse
|
43
|
Shen Z, Luo W, Tan B, Nie K, Deng M, Wu S, Xiao M, Wu X, Meng X, Tong T, Zhang C, Ma K, Liao Y, Xu J, Wang X. Roseburia intestinalis stimulates TLR5-dependent intestinal immunity against Crohn's disease. EBioMedicine 2022; 85:104285. [PMID: 36182776 PMCID: PMC9526137 DOI: 10.1016/j.ebiom.2022.104285] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/25/2022] Open
Abstract
Background Methods Findings Interpretation Funding
Collapse
|
44
|
Johnson SD, Knight LA, Kumar N, Olwenyi OA, Thurman M, Mehra S, Mohan M, Byrareddy SN. Early treatment with anti-α 4β 7 antibody facilitates increased gut macrophage maturity in SIV-infected rhesus macaques. Front Immunol 2022; 13:1001727. [PMID: 36389795 PMCID: PMC9664000 DOI: 10.3389/fimmu.2022.1001727] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/10/2022] [Indexed: 11/24/2022] Open
Abstract
Despite advances in combination antiretroviral therapy (cART), people living with HIV (PLWH) continue to experience gastrointestinal dysfunction. Infusions of anti-α4β7 monoclonal antibodies (mAbs) have been proposed to increase virologic control during simian immunodeficiency virus (SIV) infection in macaques with mixed results. Recent evidences suggested that therapeutic efficacy of vedolizumab (a humanized anti-α4β7 mAb), during inflammatory bowel diseases depends on microbiome composition, myeloid cell differentiation, and macrophage phenotype. We tested this hypothesis in SIV-infected, anti-α4β7 mAb-treated macaques and provide flow cytometric and microscopic evidence that anti-α4β7 administered to SIV-infected macaques increases the maturity of macrophage phenotypes typically lost in the small intestines during SIV disease progression. Further, this increase in mature macrophage phenotype was associated with tissue viral loads. These phenotypes were also associated with dysbiosis markers in the gut previously identified as predictors of HIV replication and immune activation in PLWH. These findings provide a novel model of anti-α4β7 efficacy offering new avenues for targeting pathogenic mucosal immune response during HIV/SIV infection.
Collapse
Affiliation(s)
- Samuel D. Johnson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Lindsey A. Knight
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Narendra Kumar
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Omalla A. Olwenyi
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Michellie Thurman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Smriti Mehra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Mahesh Mohan
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
45
|
Yang Z, Lin S, Feng W, Liu Y, Song Z, Pan G, Zhang Y, Dai X, Ding X, Chen L, Wang Y. A potential therapeutic target in traditional Chinese medicine for ulcerative colitis: Macrophage polarization. Front Pharmacol 2022; 13:999179. [PMID: 36147340 PMCID: PMC9486102 DOI: 10.3389/fphar.2022.999179] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Intestinal macrophages are the main participants of intestinal immune homeostasis and intestinal inflammation. Under different environmental stimuli, intestinal macrophages can be polarized into classical activated pro-inflammatory phenotype (M1) and alternative activated anti-inflammatory phenotype (M2). Its different polarization state is the “guide” to promoting the development and regression of inflammation. Under normal circumstances, intestinal macrophages can protect the intestine from inflammatory damage. However, under the influence of some genetic and environmental factors, the polarization imbalance of intestinal M1/M2 macrophages will lead to the imbalance in the regulation of intestinal inflammation and transform the physiological inflammatory response into pathological intestinal injury. In UC patients, the disorder of intestinal inflammation is closely related to the imbalance of intestinal M1/M2 macrophage polarization. Therefore, restoring the balance of M1/M2 macrophage polarization may be a potentially valuable therapeutic strategy for UC. Evidence has shown that traditional Chinese medicine (TCM) has positive therapeutic effects on UC by restoring the balance of M1/M2 macrophage polarization. This review summarizes the clinical evidence of TCM for UC, the vital role of macrophage polarization in the pathophysiology of UC, and the potential mechanism of TCM regulating macrophage polarization in the treatment of UC. We hope this review may provide some new enlightenment for the clinical treatment, fundamental research, and research and development of new Chinese medicine of UC.
Collapse
Affiliation(s)
- Zhihua Yang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shanshan Lin
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wanying Feng
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yangxi Liu
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihui Song
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guiyun Pan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhang Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiangdong Dai
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinya Ding
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lu Chen
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Lu Chen, ; Yi Wang,
| | - Yi Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Lu Chen, ; Yi Wang,
| |
Collapse
|
46
|
Liu X, Ren X, Zhou L, Liu K, Deng L, Qing Q, Li J, Zhi F, Li M. Tollip Orchestrates Macrophage Polarization to Alleviate Intestinal Mucosal Inflammation. J Crohns Colitis 2022; 16:1151-1167. [PMID: 35134154 DOI: 10.1093/ecco-jcc/jjac019] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/13/2021] [Accepted: 01/29/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Regulation of macrophage polarization is a promising strategy for treating inflammatory bowel disease [IBD]. Tollip is an important negative regulator of Toll-like receptor [TLR]-mediated innate immunity with downregulated expression in the colon tissues of patients with IBD. This study aimed to regulate the expression of Tollip to affect macrophage polarization. METHODS A molecular, targeted immunotherapy method was developed by linking mannose-modified trimethyl chitosan [MTC] with Tollip-expressing plasmids via ionic cross-linking, forming MTC-Tollip nanoparticles with a targeting function. MTC-Tollip selectively targeted mouse intestinal macrophages to regulate the polarization of macrophages for mucosal repair. RESULTS Orally administered MTC-Tollip significantly elevated Tollip expression in intestinal tissue. Compared with MTC-negative control [NC]-treated mice in which colitis was induced with dextran sodium sulphate [DSS], the MTC-Tollip nanoparticle-treated mice exhibited decreased body weight loss and colon shortening, lower proinflammatory cytokine expression in colon tissues, and greater mucosal barrier integrity. MTC-Tollip treatment decreased TNF-α and iNOS expression but increased CD206 and Arg-1 expression in colon tissue. Tollip overexpression in mouse peritoneal macrophages inhibited lipopolysaccharide [LPS]-induced proinflammatory cytokine production and promoted IL-4-induced M2 expression. The progression of peritoneal macrophages extracted from Tollip-/- mice confirmed the effect of Tollip on macrophage polarization. Western blots showed that Tollip overexpression attenuated the upregulation of TLR pathway-associated targets in M1 macrophages. CONCLUSIONS MTC nanoparticles can be 'intelligent' carriers in immunotherapy. The modulation of Tollip expression in macrophages may be a novel treatment approach for IBD.
Collapse
Affiliation(s)
- Xiaoming Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xingxing Ren
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lifeng Zhou
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Gastroenterology, Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Ke Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liangjun Deng
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Qing Qing
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jin Li
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fachao Zhi
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingsong Li
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
47
|
Zhang Q, Chen LH, Yang H, Fang YC, Wang SW, Wang M, Yuan QT, Wu W, Zhang YM, Liu ZJ, Nan FJ, Xie X. GPR84 signaling promotes intestinal mucosal inflammation via enhancing NLRP3 inflammasome activation in macrophages. Acta Pharmacol Sin 2022; 43:2042-2054. [PMID: 34912006 PMCID: PMC9343429 DOI: 10.1038/s41401-021-00825-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 11/16/2021] [Indexed: 02/06/2023] Open
Abstract
The putative medium-chain free fatty acid receptor GPR84 is a G protein-coupled receptor primarily expressed in myeloid cells that constitute the innate immune system, including neutrophils, monocytes, and macrophages in the periphery and microglia in the brain. The fact that GPR84 expression in leukocytes is remarkably increased under acute inflammatory stimuli such as lipopolysaccharide (LPS) and TNFα suggests that it may play a role in the development of inflammatory and fibrotic diseases. Here we demonstrate that GPR84 is highly upregulated in inflamed colon tissues of active ulcerative colitis (UC) patients and dextran sulfate sodium (DSS)-induced colitis mice. Infiltrating GPR84+ macrophages are significantly increased in the colonic mucosa of both the UC patients and the mice with colitis. Consistently, GPR84-/- mice are resistant to the development of colitis induced by DSS. GPR84 activation imposes pro-inflammatory properties in colonic macrophages through enhancing NLRP3 inflammasome activation, while the loss of GPR84 prevents the M1 polarization and properties of proinflammatory macrophages. CLH536, a novel GPR84 antagonist discovered by us, suppresses colitis by reducing the polarization and function of pro-inflammatory macrophages. These results define a unique role of GPR84 in innate immune cells and intestinal inflammation, and suggest that GPR84 may serve as a potential drug target for the treatment of UC.
Collapse
Affiliation(s)
- Qing Zhang
- grid.9227.e0000000119573309State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024 China
| | - Lin-hai Chen
- grid.9227.e0000000119573309State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Hui Yang
- grid.9227.e0000000119573309State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - You-chen Fang
- grid.9227.e0000000119573309State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Si-wei Wang
- grid.9227.e0000000119573309State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Min Wang
- grid.9227.e0000000119573309State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Qian-ting Yuan
- grid.9227.e0000000119573309State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Wei Wu
- grid.24516.340000000123704535Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai, 200072 China
| | | | - Zhan-ju Liu
- grid.24516.340000000123704535Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Tongji University, Shanghai, 200072 China
| | - Fa-jun Nan
- grid.9227.e0000000119573309State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024 China ,Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai, 264000 China
| | - Xin Xie
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China. .,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
48
|
D'Haens G, Danese S, Davies M, Watanabe M, Hibi T. A phase II, Multicentre, Randomised, Double-Blind, Placebo-controlled Study to Evaluate Safety, Tolerability, and Efficacy of Amiselimod in Patients with Moderate to Severe Active Crohn's Disease. J Crohns Colitis 2022; 16:746-756. [PMID: 34758080 DOI: 10.1093/ecco-jcc/jjab201] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Amiselimod is an oral selective S1P1 receptor modulator with potentially fewer adverse effects than fingolimod. We evaluated the safety, tolerability, and clinical efficacy of amiselimod in participants with moderate to severe active Crohn's disease. METHODS This was a phase IIa, multicentre, randomised, double-blind, parallel group, placebo-controlled study comparing amiselimod 0.4 mg with placebo over a 14-Week treatment period. The primary endpoint of the study was the proportion of participants with clinical response (Crohn's Disease activity Index [CDAI] 100) from baseline at Week 12. RESULTS A total of 180 patients were screened and 78 were randomised [40 to amiselimod 0.4 mg and 38 to placebo]. There was no significant difference in the proportion of patients achieving CDAI 100 at Week 12 on amiselimod 0.4 mg and on placebo [48.7% vs. 54.1%, respectively] (odds ratio [OR] [95% confidence interval]: 0.79 [0.31, 1.98]). The results from the secondary endpoint analyses supported the results of the primary endpoint analysis. Treatment with amiselimod 0.4 mg was generally well tolerated, with 71.8% of participants completing the 14-week treatment period. Seven participants had serious adverse events and four discontinued treatment in the amiselimod group. CONCLUSIONS Amiselimod 0.4 mg for 12 weeks was not superior to placebo for the induction of clinical response [CDAI 100] in Crohn's disease. Treatment with amiselimod 0.4 mg was generally well tolerated and no new safety concerns related to amiselimod were reported in this study.
Collapse
Affiliation(s)
- Geert D'Haens
- Inflammatory Bowel Disease Centre, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Silvio Danese
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Ospedale San Raffaele and University Vita-Salute San Raffaele, Milano, Italy
| | - Martin Davies
- Clinical Operations, Mitsubishi Tanabe Pharma Europe, London, UK
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshifumi Hibi
- Center for Advanced IBD Research and Treatment, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| |
Collapse
|
49
|
Wu J, Wu Y, Chen Y, Liu M, Yu H, Zhang Y, Wang T. Desmethylbellidifolin Attenuates Dextran Sulfate Sodium-Induced Colitis: Impact on Intestinal Barrier, Intestinal Inflammation and Gut Microbiota. PLANTA MEDICA 2022; 88:559-569. [PMID: 34098585 DOI: 10.1055/a-1506-3476] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Ulcerative colitis has been recognized as a chronic inflammatory disease predominantly disturbing the colon and rectum. Clinically, the aminosalicylates, steroids, immunosuppressants, and biological drugs are generally used for the treatment of ulcerative colitis at different stages of disease progression. However, the therapeutic efficacy of these drugs does not satisfy the patients due to the frequent drug resistance. Herein, we reported the anti-ulcerative colitis activity of desmethylbellidifolin, a xanthone isolated from Gentianella acuta, in dextran sulfate sodium-induced colitis in mice. C57BL/6 mice were treated with 2% dextran sulfate sodium in drinking water to induce acute colitis. Desmethylbellidifolin or balsalazide sodium was orally administrated once a day. Biological samples were collected for immunohistological analysis, intestinal barrier function evaluation, cytokine measurement, and gut microbiota analysis. The results revealed that desmethylbellidifolin alleviated colon shortening and body weight loss in dextran sulfate sodium-induced mice. The disease activity index was also lowered by desmethylbellidifolin after 9 days of treatment. Furthermore, desmethylbellidifolin remarkably ameliorated colonic inflammation through suppressing the expression of interleukin-6 and tumor necrosis factor-α. The intestinal epithelial barrier was strengthened by desmethylbellidifolin through increasing levels of occludin, ZO-1, and claudins. In addition, desmethylbellidifolin modulated the gut dysbiosis induced by dextran sulfate sodium. These findings suggested that desmethylbellidifolin effectively improved experimental ulcerative colitis, at least partly, through maintaining intestinal barrier integrity, inhibiting proinflammatory cytokines, and modulating dysregulated gut microbiota.
Collapse
Affiliation(s)
- Jiaqi Wu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuzheng Wu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yue Chen
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mengyang Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Haiyang Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Tao Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
50
|
Li Z, Kuang X, Chen T, Shen T, Wu J. Peptide YY 3-36 attenuates trinitrobenzene sulfonic acid-induced colitis in mice by modulating Th1/Th2 differentiation. Bioengineered 2022; 13:10144-10158. [PMID: 35443853 PMCID: PMC9161959 DOI: 10.1080/21655979.2022.2064147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Peptide YY (PYY) 3–36, the main circulatory form of PYY, plays important roles in gastrointestinal motility, secretion, and absorption. However, it is unknown whether PYY 3–36 has underlying functions in colitis. The Crohn’s disease (CD)-like mouse model in which CD is induced by trinitrobenzene sulfonic acid (TNBS) was established and utilized to investigate this potential role for PYY 3–36. The results showed that the expression of colonic mucosal PYY and PYY receptors Y1, Y2, Y4 were significantly increased in mice with TNBS-induced colitis. In vitro, PYY 3–36 remarkably inhibited the production of proinflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) from lipopolysaccharide (LPS)-induced macrophages. In vivo, a high concentration of PYY 3–36 robustly decreased the weight loss and death rate and attenuated the pathological colon tissue damage observed in mice with TNBS-induced colitis. Further studies uncovered that PYY 3–36 treatment reduced the levels of colon myeloperoxidase (MPO) and both colonic and systemic TNF-α and IL-6 observed in murine colitis. Furthermore, flow cytometric analysis showed PYY 3–36 altered the proportion of Th1/Th2 splenocytes in the disease model of colitis. Collectively, these results suggest that PYY 3–36 may be a promising candidate for the improvement of colitis, reflected by the attenuation of colon inflammatory responses observed in experimental murine colitis.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Immunology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.,Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medicine, Guizhou Medical University, Department of Medical Parasitology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xiaoyuan Kuang
- Department of Immunology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Tao Chen
- Graduate School, Zunyi Medical University, Zunyi, Guizhou, China
| | - Tao Shen
- Department of Immunology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jiahong Wu
- Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medicine, Guizhou Medical University, Department of Medical Parasitology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|