1
|
Vucetic A, Lafleur A, Côté M, Kobasa D, Chan M, Alvarez F, Piccirillo C, Dong G, Olivier M. Extracellular vesicle storm during the course of Ebola virus infection in primates. Front Cell Infect Microbiol 2023; 13:1275277. [PMID: 38035334 PMCID: PMC10684970 DOI: 10.3389/fcimb.2023.1275277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction Ebola virus (EBOV) is an RNA virus of the Filoviridae family that is responsible for outbreaks of hemorrhagic fevers in primates with a lethality rate as high as 90%. EBOV primarily targets host macrophages leading to cell activation and systemic cytokine storm, and fatal infection is associated with an inhibited interferon response, and lymphopenia. The EBOV surface glycoprotein (GP) has been shown to directly induce T cell depletion and can be secreted outside the virion via extracellular vesicles (EVs), though most studies are limited to epithelial cells and underlying mechanisms remain poorly elucidated. Methods To assess the role of GP on EBOV-induced dysregulation of host immunity, we first utilized EBOV virus-like particles (VLPs) expressing VP40 and NP either alone (Bald-VLP) or in conjunction with GP (VLP-GP) to investigate early inflammatory responses in THP-1 macrophages and in a murine model. We then sought to decipher the role of non-classical inflammatory mediators such as EVs over the course of EBOV infection in two EBOV-infected rhesus macaques by isolating and characterizing circulatory EVs throughout disease progression using size exclusion chromatography, nanoparticle tracking-analysis, and LC-MS/MS. Results While all VLPs could induce inflammatory mediators and recruit small peritoneal macrophages, pro-inflammatory cytokine and chemokine gene expression was exacerbated by the presence of GP. Further, quantification of EVs isolated from infected rhesus macaques revealed that the concentration of vesicles peaked in circulation at the terminal stage, at which time EBOV GP could be detected in host-derived exosomes. Moreover, comparative proteomics conducted across EV populations isolated from serum at various time points before and after infection revealed differences in host-derived protein content that were most significantly pronounced at the endpoint of infection, including significant expression of mediators of TLR4 signaling. Discussion These results suggest a dynamic role for EVs in the modification of disease states in the context of EBOV. Overall, our work highlights the importance of viral factors, such as the GP, and host derived EVs in the inflammatory cascade and pathogenesis of EBOV, which can be collectively further exploited for novel antiviral development.
Collapse
Affiliation(s)
- Andrea Vucetic
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Andrea Lafleur
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology and Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| | - Darwyn Kobasa
- Special Pathogen Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Mable Chan
- Special Pathogen Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Federation of Clinical Immunology (FOCiS) Centres of Excellence in Translational Immunology (CETI), Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Ciriaco Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Federation of Clinical Immunology (FOCiS) Centres of Excellence in Translational Immunology (CETI), Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - George Dong
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Martin Olivier
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Federation of Clinical Immunology (FOCiS) Centres of Excellence in Translational Immunology (CETI), Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
2
|
Chang Y, Kang P, Cui T, Guo W, Zhang W, Du P, Yi X, Guo S, Gao T, Li C, Li S. Pharmacological inhibition of demethylzeylasteral on JAK-STAT signaling ameliorates vitiligo. J Transl Med 2023; 21:434. [PMID: 37403086 DOI: 10.1186/s12967-023-04293-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 06/22/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND The activation of CD8+ T cells and their trafficking to the skin through JAK-STAT signaling play a central role in the development of vitiligo. Thus, targeting this key disease pathway with innovative drugs is an effective strategy for treating vitiligo. Natural products isolated from medicinal herbs are a useful source of novel therapeutics. Demethylzeylasteral (T-96), extracted from Tripterygium wilfordii Hook F, possesses immunosuppressive and anti-inflammatory properties. METHODS The efficacy of T-96 was tested in our mouse model of vitiligo, and the numbers of CD8+ T cells infiltration and melanocytes remaining in the epidermis were quantified using whole-mount tail staining. Immune regulation of T-96 in CD8+ T cells was evaluated using flow cytometry. Pull-down assay, mass spectrum analysis, molecular docking, knockdown and overexpression approaches were utilized to identify the target proteins of T-96 in CD8+ T cells and keratinocytes. RESULTS Here, we found that T-96 reduced CD8+ T cell infiltration in the epidermis using whole-mount tail staining and alleviated the extent of depigmentation to a comparable degree of tofacitinib (Tofa) in our vitiligo mouse model. In vitro, T-96 decreased the proliferation, CD69 membrane expression, and IFN-γ, granzyme B, (GzmB), and perforin (PRF) levels in CD8+ T cells isolated from patients with vitiligo. Pull-down assays combined with mass spectrum analysis and molecular docking showed that T-96 interacted with JAK3 in CD8+ T cell lysates. Furthermore, T-96 reduced JAK3 and STAT5 phosphorylation following IL-2 treatment. T-96 could not further reduce IFN-γ, GzmB and PRF expression following JAK3 knockdown or inhibit increased immune effectors expression upon JAK3 overexpression. Additionally, T-96 interacted with JAK2 in IFN-γ-stimulated keratinocytes, inhibiting the activation of JAK2, decreasing the total and phosphorylated protein levels of STAT1, and reducing the production and secretion of CXCL9 and CXCL10. T-96 did not significantly inhibit STAT1 and CXCL9/10 expression following JAK2 knockdown, nor did it suppress upregulated STAT1-CXCL9/10 signaling upon JAK2 overexpression. Finally, T-96 reduced the membrane expression of CXCR3, and the culture supernatants pretreated with T-96 under IFN-γ stressed keratinocytes markedly blocked the migration of CXCR3+CD8+ T cells, similarly to Tofa in vitro. CONCLUSION Our findings demonstrated that T-96 might have positive therapeutic responses to vitiligo by pharmacologically inhibiting the effector functions and skin trafficking of CD8+ T cells through JAK-STAT signaling.
Collapse
Affiliation(s)
- Yuqian Chang
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Pan Kang
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Tingting Cui
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Weinan Guo
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Weigang Zhang
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Pengran Du
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Xiuli Yi
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Sen Guo
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Tianwen Gao
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China.
| | - Shuli Li
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
3
|
Kong W, Li X, Zou M, Zhang Y, Cai H, Zhang L, Wang X. iNKT17 cells play a pathogenic role in ethinylestradiol-induced cholestatic hepatotoxicity. Arch Toxicol 2023; 97:561-580. [PMID: 36329302 DOI: 10.1007/s00204-022-03403-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
IL-17 is closely associated with inflammation in intrahepatic cholestasis (IHC). Targeting IL-17 ameliorates IHC in mice. Invariant natural killer T (iNKT) cells are predominantly enriched in the liver and they mediate drug-induced liver injury through their secreted cytokines. However, whether iNKT17 cells are involved in ethinylestradiol (EE)-induced IHC remains unclear. In the present study, the administration of EE (10 mg/kg in vivo and 6.25 μM in vitro) promoted the activation and expansion of iNKT17 cells, which contributed to a novel hepatic iNKT17/Treg imbalance. iNKT cell-deficient Jα18-/- mice and the RORγt inhibitor digoxin (20 μg) alleviated EE-induced cholestatic hepatotoxicity and downregulated the IL-17 signalling pathway. In contrast, the co-administration of EE with recombinant IL-17 (1 μg) to Jα18-/- mice induced cholestatic hepatotoxicity and increased the infiltration of hepatic neutrophils and monocytes. Importantly, the administration of IL-17-/- iNKT cells (3.5 × 105) to Jα18-/- mice resulted in the attenuation of hepatotoxicity and the recruitment of fewer hepatic neutrophils and monocytes than the adoptive transfer of wild-type iNKT cells. These results indicated that iNKT17 cells could exert pathogenic effects. The recruitment and activation of iNKT17 cells could be attributed to the high level of CXCR3 expression on their surface. CXCL10 deficiency ameliorated EE-induced cholestatic liver damage, reduced hepatic CXCR3+ iNKT cells and inhibited RORγt expression. These findings suggest that iNKT17 cells play a key role in EE-induced cholestatic liver injury via CXCR3-mediated recruitment and activation. Our study provides new insights and therapeutic targets for cholestatic diseases.
Collapse
Affiliation(s)
- Weichao Kong
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Xinyu Li
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Mengzhi Zou
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Yiying Zhang
- Division of Biosciences, University College London, London, WC1E 6BT, UK
| | - Heng Cai
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, 210009, China
| | - Luyong Zhang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, 210009, China.
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Xinzhi Wang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
4
|
Saber MM. Coexpression of PD-L1/PD-1 with CXCR3/CD36 and IL-19 Increase in Extranodal Lymphoma. J Immunol Res 2023; 2023:4556586. [PMID: 36726488 PMCID: PMC9886470 DOI: 10.1155/2023/4556586] [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: 05/04/2022] [Revised: 11/16/2022] [Accepted: 12/06/2022] [Indexed: 01/25/2023] Open
Abstract
Many studies have demonstrated that PD-L1/PD-1 signaling is an immune evasion mechanism in tumors. PD-L1/PD-1 coexpression with CXCR3/CD36 in peripheral lymphocytes in lymphoma still needs to be clarified. The current study investigated PD-L1/PD-1 coexpression with CXCR3/CD36 in circulating lymphocytes, serum IL-19 levels, and their correlation with clinical outcome and extranodal involvement in lymphoma. Subjects and Methods. The coexpression of PD-L1/PD-1 with CXCR3/CD36 on circulating lymphocytes was analyzed by flow cytometry in 78 lymphoma patients before and after therapy and in 50 healthy controls. The concentration levels of IL-19 in serum were assessed by an ELISA. Results. PD-L1 and PD-1 were expressed on circulating CXCR3+ and CD36+ lymphocytes in lymphoma and were significantly higher in patients with extranodal involvement than in lymphoma patients without extranodal involvement (P < 0.001). Elevated IL-19 levels were observed in lymphoma patients and increased significantly in extranodal involvement (P < 0.001). High percentages of PD-L1+CXCR3+ and PD-1+CXCR3+ lymphocytes were associated with high LDH levels, hepatomegaly, lymphedema, advanced tumor stage, and recurrence. Furthermore, patients with splenomegaly and generalized lymphadenopathy had high percentages of PD-L1+CXCR3+ lymphocytes. In addition, levels of PD-L1/PD-1 coexpression with CXCR3 and IL-19 were significantly associated with bone marrow, lung, and lymph vessel involvement. Further analysis revealed that high percentages of PD-L1+CD36+ and PD-1+CD36+ lymphocytes were associated with lung and bone marrow involvement. Patients with high levels of PD-L1/PD-1 coexpression with CXCR3 and IL-19 had inferior event-free survival (EFS) compared with that in lymphoma patients with low levels. EFS was decreased in patients with high percentages of PD-L1+CD36+ and PD-1+CD36+ lymphocytes. When using the receiver operating characteristic (ROC) curve, the superiority of IL-19 (area under the curve (AUC): 0.993) and PD-L1+CXCR3+% (AUC: 0.961) to PD-1+CXCR3+% (AUC: 0.805), PD-L1+CD36+% (AUC: 0.694), and PD-1+CD36+% (AUC 0.769) was evident in the diagnosis of extranodal involvement, identifying lymphoma patients with extranodal involvement from patients without extranodal involvement. Conclusions. Coexpression of PD-L1/PD-1 with CXCR3/CD36 in circulating lymphocytes and serum IL-19 levels contributes to poor prognosis and might be potential markers for extranodal involvement in lymphoma.
Collapse
Affiliation(s)
- Manal Mohamed Saber
- Department of Clinical Pathology, Faculty of Medicine, Minia University, Minia 61519, Egypt
| |
Collapse
|
5
|
Gao YN, Wang ZW, Yang X, Wang JQ, Zheng N. Aflatoxin M1 and ochratoxin A induce a competitive endogenous RNA regulatory network of intestinal immunosuppression by whole-transcriptome analysis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 854:158777. [PMID: 36115400 DOI: 10.1016/j.scitotenv.2022.158777] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/31/2022] [Accepted: 09/11/2022] [Indexed: 06/15/2023]
Abstract
Aflatoxin M1 (AFM1) and ochratoxin A (OTA) are common mycotoxins in cereal foods and milk products, and may cause serious negative impacts on human health. The intestine is crucial for immune regulation as it protects host homeostatic health from external contaminants; however, the underlying mechanisms of AFM1 and OTA mediated intestinal immunotoxicity remain unclear. In this study, whole transcriptome analysis was used to characterize BALB/c mouse intestines exposed to individual and combined AFM1 and OTA [3.0 mg/kg body weight (BW)] for 28 days to screen for key intestinal immunotoxicity-related differentially expressed mRNAs (DEmRNAs), differentially expressed microRNAs (DEmiRNAs), differentially expressed long non-coding RNAs (DElncRNAs), and associated enriched signaling pathways. Functional validation was then conducted in intestinal differentiated Caco-2 cells using different inhibitor assays to verify the accuracy of transcriptome and the importance of the key screened regulatory factors. In vivo data revealed that AFM1 and OTA exposure disrupted the intestines and exerted intestinal immunosuppression effects. When compared with AFM1, OTA had stronger intestinal toxicity in combined treatments. Further analyses of competitive endogenous RNA (ceRNA) regulatory networks in mice showed that AFM1 and OTA mediated-intestinal immunosuppression was putatively explained as follows: (i) toxins affected DEmRNAs regarding transfer and transduction mechanisms between cells (Csf1, Csf1r, Cxcl10, Cx3cr1, and Irf1), which were regulated by key DEmiRNAs (miR-106-x, miR-107-y, and miR-124-y) and the DElncRNA Rian, and (ii) toxins inhibited transforming growth factor-β-activated kinase 1 (TAK1)/I-kappaB kinase (IKK)/inhibitor of kappa Bα (IκBα)/p65 nuclear factor-κB (NF-κB) signaling phosphorylation levels, which was validated in differentiated Caco-2 cells using the TAK1 inhibitor (5Z-7-oxozeaenol). In conclusion, we evaluated the risk of co-exposure to AFM1 and OTA and associated health hazards from a whole transcriptome perspective.
Collapse
Affiliation(s)
- Ya-Nan Gao
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Zi-Wei Wang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xue Yang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jia-Qi Wang
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Milk and Milk Products Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
6
|
Chen ZL, Yin ZJ, Qiu TY, Chen J, Liu J, Zhang XY, Xu JQ. Revealing the characteristics of ZIKV infection through tissue-specific transcriptome sequencing analysis. BMC Genomics 2022; 23:697. [PMID: 36209057 PMCID: PMC9546753 DOI: 10.1186/s12864-022-08919-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 09/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recently, Zika virus (ZIKV) re-emerged in India and was potentially associated with microcephaly. However, the molecular mechanisms underlying ZIKV pathogenesis remain to be explored. RESULTS Herein, we performed a comprehensive RNA-sequencing analysis on ZIKV-infected JEG-3, U-251 MG, and HK-2 cells versus corresponding uninfected controls. Combined with a series of functional analyses, including gene annotation, pathway enrichment, and protein-protein interaction (PPI) network analysis, we defined the molecular characteristics induced by ZIKV infection in different tissues and invasion time points. Data showed that ZIKV infection and replication in each susceptible organ commonly stimulated interferon production and down-regulated metabolic-related processes. Also, tissue-specific immune responses or biological processes (BPs) were induced after ZIKV infection, including GnRH signaling pathway in JEG-3 cells, MAPK signaling pathway in U-251 MG cells, and PPAR signaling pathway in HK-2 cells. Of note, ZIKV infection induced delayed antiviral interferon responses in the placenta-derived cell lines, which potentially explains the molecular mechanism by which ZIKV replicates rapidly in the placenta and subsequential vertical transmission occurs. CONCLUSIONS Together, these data may provide a systemic insight into the pathogenesis of ZIKV infection in distinct human tissue-derived cell lines, which is likely to help develop prophylactic and therapeutic strategies against ZIKV infection.
Collapse
Affiliation(s)
- Zhi-Lu Chen
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Zuo-Jing Yin
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Tian-Yi Qiu
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Immunotherapy and Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Jian Chen
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Jian Liu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Xiao-Yan Zhang
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China. .,Department of Immunotherapy and Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Jian-Qing Xu
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China. .,Department of Immunotherapy and Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
7
|
Espinosa-Bautista F, Coronel D, Ramos-Rosillo V, Amezcua-Guerra LM. Performance analysis of Luminex and ELISA to profile serum IP-10 as a biomarker in systemic lupus erythematosus. Lupus 2022; 31:1660-1665. [PMID: 36040216 DOI: 10.1177/09612033221122978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Interferon-γ inducible protein-10 (IP-10) is a promising biomarker in systemic lupus erythematosus (SLE). The optimal quantification platform has not yet been identified. We compared the performance of bead-based multiplex assay (Luminex) and high-sensitivity enzyme-linked immunosorbent assay (hs-ELISA) for profiling serum IP-10 as a biomarker of lupus activity. METHODS A cross-sectional study was conducted on outpatients with SLE. Serum IP-10 was measured simultaneously on Luminex and hs-ELISA, and correlation between platforms was assessed. Additionally, IP-10 levels were tested against disease activity and organ involvement. RESULTS One-hundred and forty-one patients (88% women; 38 years old) were studied. Median IP-10 levels were 100.9 (125.2) pg/mL by Luminex and 156.5 (191.7) pg/mL by hs-ELISA. Correlation analysis showed Spearman's ρ = 0.621 (p < 0.0001) between Luminex and hs-ELISA. Quantification of IP-10 by Luminex showed a significant correlation (ρ = 0.198; p = 0.021) with disease activity, while this was not observed (ρ = 0.036; p = 0.683) when measured using hs-ELISA. Serum IP-10 levels were lower in quiescent patients than in those with active disease (70.8 [68.4] versus 114.3 [123.9] pg/mL; p = 0.024), with an AUC-ROC = 0.62 (p = 0.029), sensitivity = 47.9%, specificity = 77.5%, and positive likelihood ratio = 2.1. Patients with active arthritis had higher IP-10 levels than non-arthritis patients (158.1 [505.4] versus 94.1 [114.0] pg/mL; p = 0.008), with an AUC-ROC = 0.73 (p = 0.0009), sensitivity = 72.7%, specificity = 66.4%, and positive likelihood ratio = 2.1. No other type of organ involvement was identified by serum IP-10. CONCLUSIONS Luminex performs better than hs-ELISA as a quantification platform for IP-10 as it correlates with disease activity and identifies active arthritis in SLE.
Collapse
Affiliation(s)
- Fernanda Espinosa-Bautista
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico.,School of Medicine, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Dania Coronel
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico.,School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Varna Ramos-Rosillo
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico.,School of Medicine, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| | - Luis M Amezcua-Guerra
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico.,Health Care Department, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| |
Collapse
|
8
|
Brandt EF, Baues M, Wirtz TH, May JN, Fischer P, Beckers A, Schüre BC, Sahin H, Trautwein C, Lammers T, Berres ML. Chemokine CXCL10 Modulates the Tumor Microenvironment of Fibrosis-Associated Hepatocellular Carcinoma. Int J Mol Sci 2022; 23:ijms23158112. [PMID: 35897689 PMCID: PMC9329882 DOI: 10.3390/ijms23158112] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/05/2022] [Accepted: 07/09/2022] [Indexed: 01/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) constitutes a devastating health burden. Recently, tumor microenvironment-directed interventions have profoundly changed the landscape of HCC therapy. In the present study, the function of the chemokine CXCL10 during fibrosis-associated hepatocarcinogenesis was analyzed with specific focus on its impact in shaping the tumor microenvironment. C57BL/6J wild type (WT) and Cxcl10 knockout mice (Cxcl10−/−) were treated with diethylnitrosamine (DEN) and tetrachloromethane (CCl4) to induce fibrosis-associated HCCs. Cxcl10 deficiency attenuated hepatocarcinogenesis by decreasing tumor cell proliferation as well as tumor vascularization and modulated tumor-associated extracellular matrix composition. Furthermore, the genetic inactivation of Cxcl10 mediated an alteration of the tumor-associated immune response and modified chemokine/chemokine receptor networks. The DEN/CCl4-treated Cxcl10−/− mice presented with a pro-inflammatory tumor microenvironment and an accumulation of anti-tumoral immune cells in the tissue. The most striking alteration in the Cxcl10−/− tumor immune microenvironment was a vast accumulation of anti-tumoral T cells in the invasive tumor margin. In summary, our results demonstrate that CXCL10 exerts a non-redundant impact on several hallmarks of the tumor microenvironment and especially modulates the infiltration of anti-tumorigenic immune cells in HCC. In the era of microenvironment-targeted HCC therapies, interfering with CXCL10 defines a novel asset for further improvement of therapeutic strategies.
Collapse
Affiliation(s)
- Elisa F. Brandt
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (E.F.B.); (T.H.W.); (P.F.); (A.B.); (H.S.); (C.T.)
| | - Maike Baues
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, 52074 Aachen, Germany; (M.B.); (J.-N.M.); (B.-C.S.); (T.L.)
| | - Theresa H. Wirtz
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (E.F.B.); (T.H.W.); (P.F.); (A.B.); (H.S.); (C.T.)
| | - Jan-Niklas May
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, 52074 Aachen, Germany; (M.B.); (J.-N.M.); (B.-C.S.); (T.L.)
| | - Petra Fischer
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (E.F.B.); (T.H.W.); (P.F.); (A.B.); (H.S.); (C.T.)
| | - Anika Beckers
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (E.F.B.); (T.H.W.); (P.F.); (A.B.); (H.S.); (C.T.)
| | - Björn-Carsten Schüre
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, 52074 Aachen, Germany; (M.B.); (J.-N.M.); (B.-C.S.); (T.L.)
| | - Hacer Sahin
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (E.F.B.); (T.H.W.); (P.F.); (A.B.); (H.S.); (C.T.)
| | - Christian Trautwein
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (E.F.B.); (T.H.W.); (P.F.); (A.B.); (H.S.); (C.T.)
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), 52074 Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, University Hospital RWTH Aachen, 52074 Aachen, Germany; (M.B.); (J.-N.M.); (B.-C.S.); (T.L.)
| | - Marie-Luise Berres
- Department of Internal Medicine III, RWTH Aachen University, 52074 Aachen, Germany; (E.F.B.); (T.H.W.); (P.F.); (A.B.); (H.S.); (C.T.)
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), 52074 Aachen, Germany
- Correspondence:
| |
Collapse
|
9
|
Selvan GT, Gollapalli P, Shetty P, Kumari NS. Exploring key molecular signatures of immune responses and pathways associated with tuberculosis in comorbid diabetes mellitus: a systems biology approach. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2022. [DOI: 10.1186/s43088-022-00257-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Comorbid type 2 diabetes mellitus (T2DM) increases the risk for tuberculosis (TB) and its associated complications, although the pathological connections between T2DM and TB are unknown. The current research aims to identify shared molecular gene signatures and pathways that affirm the epidemiological association of T2DM and TB and afford clues on mechanistic basis of their association through integrative systems biology and bioinformatics approaches. Earlier research has found specific molecular markers linked to T2DM and TB, but, despite their importance, only offered a limited understanding of the genesis of this comorbidity. Our investigation used a network medicine method to find possible T2DM-TB molecular mediators.
Results
Functional annotation clustering, interaction networks, network cluster analysis, and network topology were part of our systematic investigation of T2DM-TB linked with 1603 differentially expressed genes (DEGs). The functional enrichment and gene interaction network analysis emphasized the importance of cytokine/chemokine signalling, T cell receptor signalling route, NF-kappa B signalling pathway and Jak-STAT signalling system. Furthermore, network analysis revealed significant DEGs such as ITGAM and STAT1, which may be necessary for T2DM-TB immune responses. Furthermore, these two genes are modulators in clusters C4 and C5, abundant in cytokine/chemokine signalling and Jak-STAT signalling pathways.
Conclusions
Our analyses highlight the role of ITGAM and STAT1 in T2DM-TB-associated pathways and advances our knowledge of the genetic processes driving this comorbidity.
Collapse
|
10
|
Liang Z, He P, Han Y, Yun CC. Survival of Stem Cells and Progenitors in the Intestine Is Regulated by LPA 5-Dependent Signaling. Cell Mol Gastroenterol Hepatol 2022; 14:129-150. [PMID: 35390517 PMCID: PMC9120264 DOI: 10.1016/j.jcmgh.2022.03.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Regeneration of the epithelium by stem cells in the intestine is supported by intrinsic and extrinsic factors. Lysophosphatidic acid (LPA), a bioactive lipid mediator, regulates many cellular functions, including cell proliferation, survival, and cytokine secretion. Here, we identify LPA5 receptor as a potent regulator of the survival of stem cells and transit-amplifying cells in the intestine. METHODS We have used genetic mouse models of conditional deletion of Lpar5, Lpar5f/f;Rosa-CreERT (Lpar5KO), and intestinal epithelial cell-specific Lpar5f/f;AhCre (Lpar5IECKO) mice. Mice were treated with tamoxifen or β-naphthoflavone to delete Lpar5 expression. Enteroids derived from these mice were used to determine the effect of Lpar5 loss on the apoptosis and proliferation of crypt epithelial cells. RESULTS Conditional loss of Lpar5 induced ablation of the intestinal mucosa, which increased morbidity of Lpar5KO mice. Epithelial regeneration was compromised with increased apoptosis and decreased proliferation of crypt epithelial cells by Lpar5 loss. Interestingly, intestinal epithelial cell-specific Lpar5 loss did not cause similar phenotypic defects in vivo. Lpar5 loss reduced intestinal stem cell marker gene expression and reduced lineage tracing from Lgr5+ ISCs. Lpar5 loss induced CXCL10 expression which exerts cytotoxic effects on intestinal stem cells and progenitors in the intestinal crypts. By co-culturing Lpar5KO enteroids with wild-type or Lpar5KO splenocytes, we demonstrated that lymphocytes protect the intestinal crypts via a LPA5-dependent suppression of CXCL10. CONCLUSIONS LPA5 is essential for the regeneration of intestinal epithelium. Our findings reveal a new finding that LPA5 regulates survival of stem cells and transit-amplifying cells in the intestine.
Collapse
Affiliation(s)
- Zhongxing Liang
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Peijian He
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Yiran Han
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - C. Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia,Correspondence Address correspondence to: Chris Yun, PhD, Division of Digestive Diseases, Emory University School of Medicine, Atlanta, GA 30324. fax: (404) 727-5767.
| |
Collapse
|
11
|
Li L, Du X, Fan G. Identifying Potential Biomarkers of Prognostic Value in Colorectal Cancer via Tumor Microenvironment Data Mining. Front Genet 2022; 12:787208. [PMID: 35251116 PMCID: PMC8890124 DOI: 10.3389/fgene.2021.787208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/16/2021] [Indexed: 12/21/2022] Open
Abstract
Colorectal cancer (CRC) is a common cancer that has increased rapidly worldwide in the past decades with a relatively high mortality rate. An increasing body of evidence has highlighted the importance of infiltrating immune and stromal cells in CRC. In this study, based on gene expression data of CRC patients in TCGA database we evaluated immune and stromal scores in tumor microenvironment using ESTIMATE method. Results showed there was potential correlation between these scores and the prognosis, and that patients with higher immune score and lower stromal score had longer survival time. We found that immune score was correlated with clinical characteristics including tumor location, tumor stage, and survival time. Specifically, the right-sided colon cancer had markedly elevated immune score, compared to left-sided colon cancer and rectal cancer. These results might be useful for understanding tumor microenvironment in colorectal cancer. Through the differential analysis we got a list of genes significantly associated with immune and stromal scores. Gene Set Enrichment and protein-protein interaction network analysis were used to further illustrate these differentially expressed genes. Finally, 15 hub genes were identified, and three (CXCL9, CXCL10 and SELL) of them were validated with favorable outcomes in CRC patients. Our result suggested that these tumor microenvironment related genes might be potential biomarkers for the prognosis of CRC.
Collapse
Affiliation(s)
- Lei Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Qingdao, BGI-Shenzhen, Qingdao, China
| | - Xiao Du
- BGI-Qingdao, BGI-Shenzhen, Qingdao, China
- BGI-Shenzhen, Shenzhen, China
- *Correspondence: Guangyi Fan, ; Xiao Du,
| | - Guangyi Fan
- BGI-Qingdao, BGI-Shenzhen, Qingdao, China
- BGI-Shenzhen, Shenzhen, China
- *Correspondence: Guangyi Fan, ; Xiao Du,
| |
Collapse
|
12
|
Qiao X, Zhang W, Zhao W. Role of CXCL10 in Spinal Cord Injury. Int J Med Sci 2022; 19:2058-2070. [PMID: 36483597 PMCID: PMC9724238 DOI: 10.7150/ijms.76694] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 11/03/2022] [Indexed: 11/24/2022] Open
Abstract
Spinal cord injury (SCI) results in acute inflammatory responses and secondary damages, including neuronal and glial cell death, axonal damage and demyelination, and blood-brain barrier (BBB) damage, eventually leading to neuronal dysfunction and other complications. C-X-C motif Chemokine Ligand 10 (CXCL10) is expressed after the injury, playing multiple roles in the development and progression of SCI. Moreover, the CXCL10 antagonist can restrict inflammatory immune responses and promote neuronal regeneration and functional recovery. In this review, we summarize the structure and biological functions of CXCL10, and the roles of the CXCL10 / CXCR3 axis in acute inflammatory responses, secondary damages, and complications during SCI, thus providing a potential theoretical basis by highlighting CXCL10 as a new potential drug target for the treatment of SCI.
Collapse
Affiliation(s)
- Xinyu Qiao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Wei Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.,Department of Pathogen Biology, Guizhou Nursing Vocational College, Guiyang, China
| | - Weijiang Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.,Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China
| |
Collapse
|
13
|
Gao ZY, Su LC, Wu QC, Sheng JE, Wang YL, Dai YF, Chen AP, He SS, Huang X, Yan GQ. Bioinformatics analyses of gene expression profile identify key genes and functional pathways involved in cutaneous lupus erythematosus. Clin Rheumatol 2021; 41:437-452. [PMID: 34553293 DOI: 10.1007/s10067-021-05913-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/05/2021] [Accepted: 09/05/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Lupus erythematosus is an autoimmune disease that causes damage to multiple organs ranging from skin lesions to systemic manifestations. Cutaneous lupus erythematosus (CLE) is a common type of lupus erythematosus (LE), but its molecular mechanisms are currently unknown. The study aimed to explore changes in the gene expression profiles and identify key genes involved in CLE, hoping to uncover its molecular mechanism and identify new targets for CLE. METHOD We analyzed the microarray dataset (GSE109248) derived from the Gene Expression Omnibus (GEO) database, which was a transcriptome profiling of CLE cutaneous lesions. The differentially expressed genes (DEGs) were identified, and the functional annotation of DEGs was performed with Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Protein-protein interaction (PPI) network was also constructed to identify hub genes involved in CLE. RESULT A total of 755 up-regulated DEGs and 405 down-regulated DEGs were identified. GO enrichment analysis showed that defense response to virus, immune response, and type I interferon signaling pathway were the most significant enrichment items in DEGs. The KEGG pathway analysis identified 51 significant enrichment pathways, which mainly included systemic lupus erythematosus, osteoclast differentiation, cytokine-cytokine receptor interaction, and primary immunodeficiency. Based on the PPI network, the study identified the top 10 hub genes involved in CLE, which were CXCL10, CCR7, FPR3, PPARGC1A, MMP9, IRF7, IL2RG, SOCS1, ISG15, and GSTM3. By comparison between subtypes, the results showed that ACLE had the least DEGs, while CCLE showed the most gene and functional changes. CONCLUSION The identified hub genes and functional pathways found in this study may expand our understanding on the underlying pathogenesis of CLE and provide new insights into potential biomarkers or targets for the diagnosis and treatment of CLE. Key Points • The bioinformatics analysis based on CLE patients and healthy controls was performed and 1160 DEGs were identified • The 1160 DEGs were mainly enriched in biological processes related to immune responses, including innate immune response, type I interferon signaling pathway, interferon-γ-mediated signaling pathway, positive regulation of T cell proliferation, regulation of immune response, antigen processing, and presentation via MHC class Ib and so on • KEGG pathway enrichment analysis indicated that DEGs were mainly enriched in several immune-related diseases and virus infection, including systemic lupus erythematosus, primary immunodeficiency, herpes simplex infection, measles, influenza A, and so on • The hub genes such as CXCL10, IRF7, MMP9, CCR7, and SOCS1 may become new markers or targets for the diagnosis and treatment of CLE.
Collapse
Affiliation(s)
- Zhen-Yu Gao
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China. .,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.
| | - Lin-Chong Su
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - Qing-Chao Wu
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - Jiao-E Sheng
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - Yun-Long Wang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - Yu-Fang Dai
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - An-Ping Chen
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - San-Shan He
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - Xia Huang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - Guo-Qing Yan
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| |
Collapse
|
14
|
Zhang Y, Wang R, Shi W, Zheng Z, Wang X, Li C, Zhang S, Zhang P. Antiviral effect of fufang yinhua jiedu (FFYH) granules against influenza A virus through regulating the inflammatory responses by TLR7/MyD88 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 275:114063. [PMID: 33813013 PMCID: PMC9759603 DOI: 10.1016/j.jep.2021.114063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 05/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fufang-Yinhua-Jiedu Granules (FFYH) optimized from a Yin-Qiao-San, as traditional Chinese medicine (TCM), was used to treat influenza and upper respiratory tract infection and was recommended for the prevention and treatment of SARS in 2003 and current COVID-19 in Anhui Province in 2020. AIM OF STUDY In the clinical studies, FFYH was very effective for the treatment of influenza, but the mechanism of action against influenza A virus remains unclear. In the present study, we investigated the antiviral effect of FFYH against influenza A virus in vitro and vivo. Moreover, the potential mechanism of FFYH against influenza A virus in vivo was investigated for the first time. MATERIALS AND METHODS CPE inhibition assay and HA assay were used to evaluate the in vitro antiviral effects of FFYH against influenza A virus H1N1, H3N2, H5N1, H7N9 and H9N2. Mice were used to evaluate the antiviral effect of FFYH in vivo with ribavirin and lianhuaqingwen as positive controls. RT-PCR was used to quantify the mRNA transcription of TNF-α, IL-6, IFN-γ, IP10, and IL-1β mRNA. ELISA was used to examine the expression of inflammatory factors such as TNF-α, IL-6, IFN-γ, IP10, and IL-1β in sera. The blood parameters were analyzed with auto hematology analyzer. Moreover, the potential mechanism of FFYH against influenza A virus in vivo was also investigated. RESULTS FFYH showed a broad-spectrum of antiviral activity against H1N1, H3N2, H5N1, H7N9, and H9N2 influenza A viruses. Furthermore, FFYH dose-dependently increased the survival rate, significantly prolonged the median survival time of mice, and markedly reduced lung injury caused by influenza A virus. Also, FFYH significantly improve the sick signs, food taken, weight loss, blood parameters, lung index, and lung pathological changes. Moreover, FFYH could markedly inhibit the inflammatory cytokine expression of TNF-α, IL-6, IFN-γ, IP10, IL-10, and IL-1β mRNA or protein via inhibition of the TLR7/MyD88/NF-κB signaling pathway in vivo. CONCLUSION FFYH not only showed a broad-spectrum of anti-influenza virus activity in vitro, but also exhibited a significant protective effect against lethal influenza virus infection in vivo. Furthermore, our results indicated that the in vivo antiviral effect of FFYH against influenza virus may be attributed to suppressing the expression of inflammatory cytokines via regulating the TLR7/MyD88/NF-κB signaling pathway. These findings provide evidence for the clinical treatment of influenza A virus infection with FFYH.
Collapse
Affiliation(s)
- Yuqian Zhang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Ronghua Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Weiqing Shi
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
| | - Zhihui Zheng
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Xiaoquan Wang
- College of Veterinary Medicine & Jiangsu Provincial Key Laboratory of Human Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Cheng Li
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Shuofeng Zhang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Pinghu Zhang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, China; College of Veterinary Medicine & Jiangsu Provincial Key Laboratory of Human Zoonosis, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
15
|
Chinnapaka S, Yang KS, Samadi Y, Epperly MW, Hou W, Greenberger JS, Ejaz A, Rubin JP. Allogeneic adipose-derived stem cells mitigate acute radiation syndrome by the rescue of damaged bone marrow cells from apoptosis. Stem Cells Transl Med 2021; 10:1095-1114. [PMID: 33724714 PMCID: PMC8235137 DOI: 10.1002/sctm.20-0455] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Acute radiation syndrome (ARS) is the radiation toxicity that can affect the hematopoietic, gastrointestinal, and nervous systems upon accidental radiation exposure within a short time. Currently, there are no effective and safe approaches to treat mass population exposure to ARS. Our study aimed to evaluate the therapeutic potential of allogeneic adipose‐derived stem cells (ASCs) for total body irradiation (TBI)‐induced ARS and understand the underlying mitigation mechanism. We employed 9.25 Gy TBI dose to C57BL/6 mice and studied the effect of allogeneic ASCs on mice survival and regeneration of the hematopoietic system. Our results indicate that intraperitoneal‐injected ASCs migrated to the bone marrow, rescued hematopoiesis, and improved the survival of irradiated mice. Our transwell coculture results confirmed the migration of ASCs to irradiated bone marrow and rescue hematopoietic activity. Furthermore, contact coculture of ASCs improved the survival and hematopoiesis of irradiated bone marrow in vitro. Irradiation results in DNA damage, upregulation of inflammatory signals, and apoptosis in bone marrow cells, while coculture with ASCs reduces apoptosis via activation of DNA repair and the antioxidation system. Upon exposure to irradiated bone marrow cells, ASCs secrete prosurvival and hematopoietic factors, such as GM‐CSF, MIP1α, MIP1β, LIX, KC, 1P‐10, Rantes, IL‐17, MCSF, TNFα, Eotaxin, and IP‐10, which reduces oxidative stress and rescues damaged bone marrow cells from apoptosis. Our findings suggest that allogeneic ASCs therapy is effective in mitigating TBI‐induced ARS in mice and may be beneficial for clinical adaptation to treat TBI‐induced toxicities. Further studies will help to advocate the scale‐up and adaptation of allogeneic ASCs as the radiation countermeasure.
Collapse
Affiliation(s)
- Somaiah Chinnapaka
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Katherine S Yang
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yasamin Samadi
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael W Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Wen Hou
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Joel S Greenberger
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - J Peter Rubin
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
16
|
Lu L, Wei R, Prats-Ejarque G, Goetz M, Wang G, Torrent M, Boix E. Human RNase3 immune modulation by catalytic-dependent and independent modes in a macrophage-cell line infection model. Cell Mol Life Sci 2021; 78:2963-2985. [PMID: 33226440 PMCID: PMC8004517 DOI: 10.1007/s00018-020-03695-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 09/21/2020] [Accepted: 10/31/2020] [Indexed: 02/06/2023]
Abstract
The human RNase3 is a member of the RNaseA superfamily involved in host immunity. RNase3 is expressed by leukocytes and shows broad-spectrum antimicrobial activity. Together with a direct antimicrobial action, RNase3 exhibits immunomodulatory properties. Here, we have analysed the transcriptome of macrophages exposed to the wild-type protein and a catalytic-defective mutant (RNase3-H15A). The analysis of differently expressed genes (DEGs) in treated THP1-derived macrophages highlighted a common pro-inflammatory "core-response" independent of the protein ribonucleolytic activity. Network analysis identified the epidermal growth factor receptor (EGFR) as the main central regulatory protein. Expression of selected DEGs and MAPK phosphorylation were inhibited by an anti-EGFR antibody. Structural analysis suggested that RNase3 activates the EGFR pathway by direct interaction with the receptor. Besides, we identified a subset of DEGs related to the protein ribonucleolytic activity, characteristic of virus infection response. Transcriptome analysis revealed an early pro-inflammatory response, not associated to the protein catalytic activity, followed by a late activation in a ribonucleolytic-dependent manner. Next, we demonstrated that overexpression of macrophage endogenous RNase3 protects the cells against infection by Mycobacterium aurum and the human respiratory syncytial virus. Comparison of cell infection profiles in the presence of Erlotinib, an EGFR inhibitor, revealed that the receptor activation is required for the antibacterial but not for the antiviral protein action. Moreover, the DEGs related and unrelated to the protein catalytic activity are associated to the immune response to bacterial and viral infection, respectively. We conclude that RNase3 modulates the macrophage defence against infection in both catalytic-dependent and independent manners.
Collapse
Affiliation(s)
- Lu Lu
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Vallès, Spain
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - RanLei Wei
- Center of Precision Medicine and Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Guillem Prats-Ejarque
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Maria Goetz
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Gang Wang
- Center of Precision Medicine and Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Marc Torrent
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Ester Boix
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Vallès, Spain.
| |
Collapse
|
17
|
La Sala L, Crestani M, Garavelli S, de Candia P, Pontiroli AE. Does microRNA Perturbation Control the Mechanisms Linking Obesity and Diabetes? Implications for Cardiovascular Risk. Int J Mol Sci 2020; 22:ijms22010143. [PMID: 33375647 PMCID: PMC7795227 DOI: 10.3390/ijms22010143] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 02/06/2023] Open
Abstract
Metabolic disorders such as obesity and type 2 diabetes (T2D) are considered the major risk factors for the development of cardiovascular diseases (CVD). Although the pathological mechanisms underlying the mutual development of obesity and T2D are difficult to define, a better understanding of the molecular aspects is of utmost importance to identify novel therapeutic targets. Recently, a class of non-coding RNAs, called microRNAs (miRNAs), are emerging as key modulators of metabolic abnormalities. There is increasing evidence supporting the role of intra- and extracellular miRNAs as determinants of the crosstalk between adipose tissues, liver, skeletal muscle and other organs, triggering the paracrine communication among different tissues. miRNAs may be considered as risk factors for CVD due to their correlation with cardiovascular events, and in particular, may be related to the most prominent risk factors. In this review, we describe the associations observed between miRNAs expression levels and the most common cardiovascular risk factors. Furthermore, we sought to depict the molecular aspect of the interplay between obesity and diabetes, investigating the role of microRNAs in the interorgan crosstalk. Finally, we discussed the fascinating hypothesis of the loss of protective factors, such as antioxidant defense systems regulated by such miRNAs.
Collapse
Affiliation(s)
- Lucia La Sala
- Laboratory of Cardiovascular and Dysmetabolic Disease, IRCCS MultiMedica, 20138 Milan, Italy;
- Correspondence:
| | - Maurizio Crestani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Silvia Garavelli
- Laboratorio di Immunologia, Istituto per l’Endocrinologia e l’Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy;
| | - Paola de Candia
- Laboratory of Cardiovascular and Dysmetabolic Disease, IRCCS MultiMedica, 20138 Milan, Italy;
| | - Antonio E. Pontiroli
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20142 Milan, Italy;
| |
Collapse
|
18
|
Mikolajczyk TP, Szczepaniak P, Vidler F, Maffia P, Graham GJ, Guzik TJ. Role of inflammatory chemokines in hypertension. Pharmacol Ther 2020; 223:107799. [PMID: 33359600 DOI: 10.1016/j.pharmthera.2020.107799] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Hypertension is associated with immune cells activation and their migration into the kidney, vasculature, heart and brain. These inflammatory mechanisms are critical for blood pressure regulation and mediate target organ damage, creating unique novel targets for pharmacological modulation. In response to angiotensin II and other pro-hypertensive stimuli, the expression of several inflammatory chemokines and their receptors is increased in the target organs, mediating homing of immune cells. In this review, we summarize the contribution of key inflammatory chemokines and their receptors to increased accumulation of immune cells in target organs and effects on vascular dysfunction, remodeling, oxidative stress and fibrosis, all of which contribute to blood pressure elevation. In particular, the role of CCL2, CCL5, CXCL8, CXCL9, CXCL10, CXCL11, CXCL16, CXCL1, CX3CL1, XCL1 and their receptors in the context of hypertension is discussed. Recent studies have tested the efficacy of pharmacological or genetic targeting of chemokines and their receptors on the development of hypertension. Promising results indicate that some of these pathways may serve as future therapeutic targets to improve blood pressure control and prevent target organ consequences including kidney failure, heart failure, atherosclerosis or cognitive impairment.
Collapse
Affiliation(s)
- Tomasz P Mikolajczyk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland; Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Piotr Szczepaniak
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Francesca Vidler
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Pasquale Maffia
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK; BHF Centre for Excellence Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Gerard J Graham
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Tomasz J Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland; BHF Centre for Excellence Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
19
|
Gao J, Wu L, Wang S, Chen X. Role of Chemokine (C-X-C Motif) Ligand 10 (CXCL10) in Renal Diseases. Mediators Inflamm 2020; 2020:6194864. [PMID: 32089645 PMCID: PMC7025113 DOI: 10.1155/2020/6194864] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 12/02/2019] [Accepted: 12/23/2019] [Indexed: 12/31/2022] Open
Abstract
Chemokine C-X-C ligand 10 (CXCL10), also known as interferon-γ-inducible protein 10 (IP-10), exerts biological function mainly through binding to its specific receptor, CXCR3. Studies have shown that renal resident mesangial cells, renal tubular epithelial cells, podocytes, endothelial cells, and infiltrating inflammatory cells express CXCL10 and CXCR3 under inflammatory conditions. In the last few years, strong experimental and clinical evidence has indicated that CXCL10 is involved in the development of renal diseases through the chemoattraction of inflammatory cells and facilitation of cell growth and angiostatic effects. In addition, CXCL10 has been shown to be a significant biomarker of disease severity, and it can be used as a prognostic indicator for a variety of renal diseases, such as renal allograft dysfunction and lupus nephritis. In this review, we summarize the structures and biological functions of CXCL10 and CXCR3, focusing on the important role of CXCL10 in the pathogenesis of kidney disease, and provide a theoretical basis for CXCL10 as a potential biomarker and therapeutic target in human kidney disease.
Collapse
Affiliation(s)
- Jie Gao
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing 100853, China
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jingwu Road 324, Jinan 250000, China
| | - Lingling Wu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing 100853, China
| | - Siyang Wang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing 100853, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing 100853, China
| |
Collapse
|
20
|
Zhang X, Fan L, Wu J, Xu H, Leung WY, Fu K, Wu J, Liu K, Man K, Yang X, Han J, Ren J, Yu J. Macrophage p38α promotes nutritional steatohepatitis through M1 polarization. J Hepatol 2019; 71:163-174. [PMID: 30914267 DOI: 10.1016/j.jhep.2019.03.014] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 03/11/2019] [Accepted: 03/14/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS p38 mitogen-activated protein kinases are important inflammatory factors. p38α alteration has been implicated in both human and mouse inflammatory disease models. Therefore, we aimed to characterize the cell type-specific role of p38α in non-alcoholic steatohepatitis (NASH). METHODS Human liver tissues were obtained from 27 patients with non-alcoholic fatty liver disease (NAFLD) and 20 control individuals. NASH was established and compared between hepatocyte-specific p38α knockout (p38αΔHep), macrophage-specific p38α knockout (p38αΔMΦ) and wild-type (p38αfl/fl) mice fed with high-fat diet (HFD), high-fat/high-cholesterol diet (HFHC), or methionine-and choline-deficient diet (MCD). p38 inhibitors were administered to HFHC-fed wild-type mice for disease treatment. RESULTS p38α was significantly upregulated in the liver tissues of patients with NAFLD. Compared to p38αfl/fl littermates, p38αΔHep mice developed significant nutritional steatohepatitis induced by HFD, HFHC or MCD. Meanwhile, p38αΔMΦ mice exhibited less severe steatohepatitis and insulin resistance than p38αfl/fl mice in response to a HFHC or MCD. The effect of macrophage p38α in promoting steatohepatitis was mediated by the induction of pro-inflammatory factors (CXCL2, IL-1β, CXCL10 and IL-6) secreted by M1 macrophages and associated signaling pathways. p38αΔMΦ mice exhibited M2 anti-inflammatory polarization as demonstrated by increased CD45+F4/80+CD11b+CD206+ M2 macrophages and enhanced arginase activity in liver tissues. Primary hepatocytes from p38αΔMΦ mice showed decreased steatosis and inflammatory damage. In a co-culture system, p38α deleted macrophages attenuated steatohepatitic changes in hepatocytes through decreased secretion of pro-inflammatory cytokines (TNF-α, CXCL10 and IL-6), which mediate M1 macrophage polarization in p38αΔMΦ mice. Restoration of TNF-α, CXCL10 or IL-6 induced lipid accumulation and inflammatory responses in p38αfl/fl hepatocytes co-cultured with p38αΔMΦ macrophages. Moreover, pharmacological p38 inhibitors suppressed HFHC-induced steatohepatitis. CONCLUSIONS Macrophage p38α promotes the progression of steatohepatitis by inducing pro-inflammatory cytokine secretion and M1 polarization. p38 inhibition protects against steatohepatitis. LAY SUMMARY: p38 mitogen-activated protein kinases are important inflammatory factors. In the present study, we demonstrated that p38α is upregulated in liver tissues of patients with non-alcoholic fatty liver diseases. Genetic deletion of p38α in macrophages led to ameliorated nutritional steatohepatitis in mice through decreased pro-inflammatory cytokine secretion and increased M2 macrophage polarization.
Collapse
Affiliation(s)
- Xiang Zhang
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Lina Fan
- Institute for Microbial Ecology, School of Medicine, Xiamen University, Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Hongzhi Xu
- Institute for Microbial Ecology, School of Medicine, Xiamen University, Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Wing Yan Leung
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Kaili Fu
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Jingtong Wu
- Institute for Microbial Ecology, School of Medicine, Xiamen University, Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Ken Liu
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong; Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Kwan Man
- Department of Surgery, LKS Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Xiaoyong Yang
- Section of Comparative Medicine and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, United States
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Jianlin Ren
- Institute for Microbial Ecology, School of Medicine, Xiamen University, Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, China.
| | - Jun Yu
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
21
|
Fang J, Wang C, Shen C, Shan J, Wang X, Liu L, Fan Y. The Expression of CXCL10/CXCR3 and Effect of the Axis on the Function of T Lymphocyte Involved in Oral Lichen Planus. Inflammation 2018; 42:799-810. [PMID: 30467622 DOI: 10.1007/s10753-018-0934-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The etiology of oral lichen planus (OLP) is still not clear. The purpose of this study was to explore the role of CXC chemokine receptor 3(CXCR3) and its ligand CXC motif chemokine 10(CXCL10) in the pathogenesis of OLP. We examined the expression of CXCR3 and CXCL10 in OLP patients and healthy controls by quantitative real-time PCR, Western blotting, ELISAs, and immunohistochemistry, respectively. Moreover, we detected the effects of CXCL10/CXCR3 axis on T lymphocyte migration, proliferation and apoptosis by Transwell assays, CCK8 assays, and flow cytometry. We found that the expression of CXCR3 and CXCL10 was significantly increased in OLP patients. In addition, T lymphocyte migration rate of CXCL10 stimulation group was significantly higher than that of control and CXCR3 antagonist groups. After antagonizing CXCR3, the migration ability of T lymphocytes was significantly decreased, and regardless of whether CXCL10 was added in the upper chamber culture medium, the number of migrating cells was similar. The addition of CXCL10 stimulant could stimulate the proliferation of T lymphocytes, but there was no significant difference compared with control group. After antagonizing CXCR3, the proliferation rate of T lymphocytes was significantly reduced. However, there were no significant differences in the apoptosis rates of T lymphocytes between CXCL10 stimulation group, antagonist CXCR3 group, and control group. Due to the change of expression in CXCR3 and CXCL10, and its interaction in mediating the directional migration of peripheral blood T lymphocytes, affecting the proliferation of T lymphocytes, it suggests that CXCL10/CXCR3 axis may be related to the immune mechanism of OLP.
Collapse
Affiliation(s)
- Jiaxiang Fang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Chen Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Chen Shen
- Department of Special outpatient service, Hangzhou West Dental Hospital, Hangzhou, 310012, China
| | - Jing Shan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Xuewei Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Lin Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China
| | - Yuan Fan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China.
- Department of Oral Medicine, Affiliated Hospital of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, 210029, China.
| |
Collapse
|
22
|
Dubnika A, Manoukian MA, Mohammadi MR, Parekh MB, Gurjarpadhye AA, Inayathullah M, Dubniks V, Lakey JR, Rajadas J. Cytokines as therapeutic agents and targets in heart disease. Cytokine Growth Factor Rev 2018; 43:54-68. [DOI: 10.1016/j.cytogfr.2018.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/01/2018] [Accepted: 08/13/2018] [Indexed: 02/02/2023]
|
23
|
Skinner D, Marro BS, Lane TE. Chemokine CXCL10 and Coronavirus-Induced Neurologic Disease. Viral Immunol 2018; 32:25-37. [PMID: 30109979 DOI: 10.1089/vim.2018.0073] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Chemokines (chemotactic cytokines) are involved in a wide variety of biological processes. Following microbial infection, there is often robust chemokine signaling elicited from infected cells, which contributes to both innate and adaptive immune responses that control growth of the invading pathogen. Infection of the central nervous system (CNS) by the neuroadapted John Howard Mueller (JHM) strain of mouse hepatitis virus (JHMV) provides an excellent example of how chemokines aid in host defense as well as contribute to disease. Intracranial inoculation of the CNS of susceptible mice with JHMV results in an acute encephalomyelitis characterized by widespread dissemination of virus throughout the parenchyma. Virus-specific T cells are recruited to the CNS, and control viral replication through release of antiviral cytokines and cytolytic activity. Sterile immunity is not acquired, and virus will persist primarily in white matter tracts leading to chronic neuroinflammation and demyelination. Chemokines are expressed and contribute to defense as well as chronic disease by attracting targeted populations of leukocytes to the CNS. The T cell chemoattractant chemokine CXCL10 (interferon-inducible protein 10 kDa, IP-10) is prominently expressed in both stages of disease, and serves to attract activated T and B lymphocytes expressing CXC chemokine receptor 3 (CXCR3), the receptor for CXCL10. Functional studies that have blocked expression of either CXCL10 or CXCR3 illuminate the important role of this signaling pathway in host defense and neurodegeneration in a model of viral-induced neurologic disease.
Collapse
Affiliation(s)
- Dominic Skinner
- 1 Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Brett S Marro
- 2 Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California
| | - Thomas E Lane
- 1 Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah.,3 Immunology, Inflammation and Infectious Disease Initiative, University of Utah School of Medicine, Salt Lake City, Utah.,4 Neuroscience Initiative, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
24
|
Chu H, Jia B, Qiu X, Pan J, Sun X, Wang Z, Zhao J. Investigation of proliferation and migration of tongue squamous cell carcinoma promoted by three chemokines, MIP-3α, MIP-1β, and IP-10. Onco Targets Ther 2017; 10:4193-4203. [PMID: 28919775 PMCID: PMC5587132 DOI: 10.2147/ott.s132855] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The aim of this work was to investigate the role of chemokines in proliferation and migration of tongue squamous cell carcinoma (TSCC). Out of the 80 cytokines surveyed by a human cytokine antibody array, three chemokines, macrophage inflammatory protein-3α (MIP-3α), macrophage inflammatory protein-1β (MIP-1β), and interferon gamma-induced protein 10 (IP-10), showed elevated expression in TSCC cells (CAL-27 and UM-1), compared to the oral mucosal epithelial cells. Immunohistochemistry confirmed the high level of expression of MIP-3α in the TSCC tissues, especially in the high clinical stages. Furthermore, Western blot and immunofluorescence staining indicated that C-C chemokine receptor type 5, C-C chemokine receptor type 6, and C-X-C motif chemokine receptor 3, which are the receptors for MIP-3α, MIP-1β, and IP-10, respectively, were expressed in the TSCC cells. Viability assay showed MIP-3α, MIP-1β, and IP-10 led to the proliferation of the CAL-27 cells. Interestingly, MIP-1β and IP-10 also induced apoptosis in the TSCC cells. Transwell invasion assay showed MIP-3α and IP-10 could increase the invasive capability of TSCC cells; consistently, the enzymatic activities of matrix metalloproteinase-2 and matrix metalloproteinase-9 increased in the MIP-3α- and IP-10-treated cells. In summary, our results indicate the expression of MIP-3α, MIP-1β, and IP-10 increased in the TSCC cells. The elevated expression of MIP-3α and IP-10 promoted proliferation and migration of TSCC. These chemokines, along with their receptors, could be potential biomarkers and therapeutic targets for TSCC, especially for those in the high clinical stages.
Collapse
Affiliation(s)
- Hongxing Chu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoling Qiu
- Department of Endodontology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Pan
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiang Sun
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiping Wang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianjiang Zhao
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
25
|
Pandya JM, Lundell AC, Andersson K, Nordström I, Theander E, Rudin A. Blood chemokine profile in untreated early rheumatoid arthritis: CXCL10 as a disease activity marker. Arthritis Res Ther 2017; 19:20. [PMID: 28148302 PMCID: PMC5289001 DOI: 10.1186/s13075-017-1224-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/06/2017] [Indexed: 12/29/2022] Open
Abstract
Background We have recently analyzed the profile of T-cell subtypes based on chemokine receptor expression in blood from untreated early rheumatoid arthritis (ueRA) patients compared to healthy controls (HC). Here, we compared the levels of the respective chemokines in blood plasma of ueRA patients with those of HC. We also studied the association of chemokine levels with the proportions of circulating T-cell subsets and the clinical disease activity. Methods Peripheral blood was obtained from 43 patients with ueRA satisfying the ACR 2010 criteria and who had not received any disease-modifying anti-rheumatic drugs (DMARD) or prednisolone, and from 14 sex- and age-matched HC. Proportions of T helper cells in blood, including Th0, Th1, Th2, Th17, Th1Th17, TFh, and regulatory T cells, were defined by flow cytometry. Fifteen chemokines, including several CXCL and CCL chemokines related to the T-cell subtypes as well as to other major immune cells, were measured in blood plasma using flow cytometry bead-based immunoassay or ELISA. Clinical disease activity in patients was evaluated by assessing the following parameters: Disease Activity Score in 28 joints (DAS28), Clinical Disease Activity Index (CDAI), swollen joint counts (SJC), tender joint counts (TJC), C-reactive protein (CRP), and erythrocyte sedimentation rate (ESR). The data were analyzed using multivariate factor analyses followed by univariate analyses. Results Multivariate discriminant analysis showed that patients with ueRA were separated from HC based on the blood plasma chemokine profile. The best discriminators were CXCL9, CXCL10, CXCL13, CCL4, and CCL22, which were significantly higher in ueRA compared to HC in univariate analyses. Among the chemokines analyzed, only CXCL10 correlated with multiple disease activity measures, including DAS28-CRP, DAS28-ESR, CDAI, SJC in 66 joints, CRP, and ESR. Conclusions High circulating levels of CXCL10 in the plasma of ueRA patients and the association with the clinical disease activity suggests that CXCL10 may serve as a disease activity marker in early rheumatoid arthritis. Electronic supplementary material The online version of this article (doi:10.1186/s13075-017-1224-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jayesh M Pandya
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy of University of Gothenburg, Box 480, S-405 30, Gothenburg, Sweden.
| | - Anna-Carin Lundell
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy of University of Gothenburg, Box 480, S-405 30, Gothenburg, Sweden
| | - Kerstin Andersson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy of University of Gothenburg, Box 480, S-405 30, Gothenburg, Sweden
| | - Inger Nordström
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy of University of Gothenburg, Box 480, S-405 30, Gothenburg, Sweden
| | - Elke Theander
- Department of Rheumatology, Skåne University Hospital Lund and Malmö, Lund University, Lund, Sweden
| | - Anna Rudin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy of University of Gothenburg, Box 480, S-405 30, Gothenburg, Sweden.
| |
Collapse
|
26
|
C-X-C motif chemokine 10 in non-alcoholic steatohepatitis: role as a pro-inflammatory factor and clinical implication. Expert Rev Mol Med 2016; 18:e16. [PMID: 27669973 DOI: 10.1017/erm.2016.16] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease. Non-alcoholic steatohepatitis (NASH) is a more severe form of NAFLD and causes subsequent pathological changes including cirrhosis and hepatocellular carcinoma. Inflammation is the key pathological change in NASH and involves a series of cytokines and chemokines. The C-X-C motif chemokine 10 (CXCL10), which is known as a pro-inflammation chemokine, was recently proven to play a pivotal role in the pathogenesis of NASH. Hepatic CXCL10 is mainly secreted by hepatocytes and liver sinusoidal endothelium. By binding to its specific receptor CXCR3, CXCL10 recruits activated CXCR3+ T lymphocytes and macrophages to parenchyma and promotes inflammation, apoptosis and fibrosis. The circulating CXCL10 level correlates with the severity of lobular inflammation and is an independent risk factor for NASH patients. Thus, CXCL10 may be both a potential prognostic tool and a therapeutic target for the treatment of patients with NASH. The aim of this review is to highlight the growing advances in basic knowledge and clinical interest of CXCL10 in NASH to propagate new insights into novel pharmacotherapeutic avenues.
Collapse
|
27
|
Haas R, Smith J, Rocher-Ros V, Nadkarni S, Montero-Melendez T, D’Acquisto F, Bland EJ, Bombardieri M, Pitzalis C, Perretti M, Marelli-Berg FM, Mauro C. Lactate Regulates Metabolic and Pro-inflammatory Circuits in Control of T Cell Migration and Effector Functions. PLoS Biol 2015; 13:e1002202. [PMID: 26181372 PMCID: PMC4504715 DOI: 10.1371/journal.pbio.1002202] [Citation(s) in RCA: 508] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 06/16/2015] [Indexed: 12/24/2022] Open
Abstract
Lactate has long been considered a “waste” by-product of cell metabolism, and it accumulates at sites of inflammation. Recent findings have identified lactate as an active metabolite in cell signalling, although its effects on immune cells during inflammation are largely unexplored. Here we ask whether lactate is responsible for T cells remaining entrapped in inflammatory sites, where they perpetuate the chronic inflammatory process. We show that lactate accumulates in the synovia of rheumatoid arthritis patients. Extracellular sodium lactate and lactic acid inhibit the motility of CD4+ and CD8+ T cells, respectively. This selective control of T cell motility is mediated via subtype-specific transporters (Slc5a12 and Slc16a1) that we find selectively expressed by CD4+ and CD8+ subsets, respectively. We further show both in vitro and in vivo that the sodium lactate-mediated inhibition of CD4+ T cell motility is due to an interference with glycolysis activated upon engagement of the chemokine receptor CXCR3 with the chemokine CXCL10. In contrast, we find the lactic acid effect on CD8+ T cell motility to be independent of glycolysis control. In CD4+ T helper cells, sodium lactate also induces a switch towards the Th17 subset that produces large amounts of the proinflammatory cytokine IL-17, whereas in CD8+ T cells, lactic acid causes the loss of their cytolytic function. We further show that the expression of lactate transporters correlates with the clinical T cell score in the synovia of rheumatoid arthritis patients. Finally, pharmacological or antibody-mediated blockade of subtype-specific lactate transporters on T cells results in their release from the inflammatory site in an in vivo model of peritonitis. By establishing a novel role of lactate in control of proinflammatory T cell motility and effector functions, our findings provide a potential molecular mechanism for T cell entrapment and functional changes in inflammatory sites that drive chronic inflammation and offer targeted therapeutic interventions for the treatment of chronic inflammatory disorders. High levels of lactate that accumulate in chronic inflammatory sites can trigger unfavorable responses in infiltrating T cells; reducing T cells' sensitivity to lactate might offer therapeutic solutions to chronic inflammatory disorders. Acidity is a feature of inflammatory sites such as arthritic synovia, atherosclerotic plaques, and tumor microenvironments and results in part from the accumulation of lactate as a product of glycolysis under hypoxic conditions. Recently it has emerged that lactate may be more than just a bystander and might act to modulate the immune-inflammatory response. Here we report just such activity: lactate inhibits T cell motility by interfering with glycolysis that is required for T cells to migrate, it causes T cells to produce higher amounts of the proinflammatory cytokine IL-17, and it triggers loss of cytolytic activity. These phenomena are hallmark features of T cells in chronic inflammatory infiltrates. The functional changes depend on the expression of specific lactate transporters by different subsets of T cells, namely the sodium lactate transporter Slc5a12 in CD4+ T cells and the lactic acid transporter Slc16a1 in CD8+ T cells. We propose that T cells entering inflammatory sites sense high concentrations of lactate via their specific transporters. Loss of motility leads to their entrapment at the site, where through their increased production of inflammatory cytokines yet decreased cytolytic capacity, they add detrimentally to chronic inflammation. Targeting lactate transporters and/or metabolic pathways on T cells could deliver novel, invaluable therapeutics for the treatment of widespread chronic inflammatory disorders.
Collapse
Affiliation(s)
- Robert Haas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Joanne Smith
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Vidalba Rocher-Ros
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Suchita Nadkarni
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Trinidad Montero-Melendez
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Fulvio D’Acquisto
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Elliot J. Bland
- Queen Mary Innovation Ltd, Queen Mary University of London, London, United Kingdom
| | - Michele Bombardieri
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Costantino Pitzalis
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Federica M. Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Claudio Mauro
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
- * E-mail:
| |
Collapse
|
28
|
Gao J, Gao J, Qian L, Wang X, Wu M, Zhang Y, Ye H, Zhu S, Yu Y, Han W. Activation of p38-MAPK by CXCL4/CXCR3 axis contributes to p53-dependent intestinal apoptosis initiated by 5-fluorouracil. Cancer Biol Ther 2014; 15:982-91. [PMID: 24800927 DOI: 10.4161/cbt.29114] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Chemotherapy-induced mucositis (CIM) is a major does limiting side-effect of chemoagents such as 5-fluorouracil (5-FU). Molecules involved in this disease process are still not fully understood. We proposed that the homeostatically regulated genes during CIM may participate in the disease. A cluster of such genes were previously identified by expression gene-array from the mouse jejunum in 5-FU-induced mucositis model. Here, we report that CXCL4 is such a homeostatically regulated gene and serves as a new target for the antibody treatment of CIM. CXCL4 and its receptor CXCR3 were confirmed at both the gene and protein levels to be homeostatically regulated during 5-FU-induced mucositis. Using of CXCL4 neutralizing monoclonal antibody (CXCL4mab) decreased the incidence, severity, and duration of the chemotherapy-induced diarrhea, the major symptom of CIM, in a 5-FU mouse CIM model. Mechanistically, CXCL4mab reduced the apoptosis of the crypt epithelia by suppression of the 5-FU-induced expression of p53 and Bax through its receptor CXCR3. The downstream signaling pathway of CXCL4 in activation of the epithelial apoptosis was identified in an intestinal epithelial cell line (IEC-6). CXCL4 activated the phosphorylation of p38 MAPK, which mediated the stimulated expression of p53 and Bax, and resulted in the ultimate activation of Caspase-8, -9, and -3. Taken together, activation of CXCL4 expression by 5-FU in mice participates in 5-FU-induced intestinal mucositis through upregulation of p53 via activation of p38-MAPK, and CXCL4mab is potentially beneficial in preventing CIM in the intestinal tract.
Collapse
Affiliation(s)
- Jing Gao
- Laboratory of Regeneromics; School of Pharmacy; Shanghai Jiao Tong University; Shanghai, PR China
| | - Jin Gao
- Laboratory of Regeneromics; School of Pharmacy; Shanghai Jiao Tong University; Shanghai, PR China
| | - Lan Qian
- Laboratory of Regeneromics; School of Pharmacy; Shanghai Jiao Tong University; Shanghai, PR China
| | - Xia Wang
- Laboratory of Regeneromics; School of Pharmacy; Shanghai Jiao Tong University; Shanghai, PR China
| | - Mingyuan Wu
- Laboratory of Regeneromics; School of Pharmacy; Shanghai Jiao Tong University; Shanghai, PR China
| | - Yang Zhang
- Laboratory of Regeneromics; School of Pharmacy; Shanghai Jiao Tong University; Shanghai, PR China
| | - Hao Ye
- Laboratory of Regeneromics; School of Pharmacy; Shanghai Jiao Tong University; Shanghai, PR China
| | - Shunying Zhu
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology; School of Agriculture and Biology; Shanghai Jiao Tong University; Shanghai, PR China
| | - Yan Yu
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology; School of Agriculture and Biology; Shanghai Jiao Tong University; Shanghai, PR China
| | - Wei Han
- Laboratory of Regeneromics; School of Pharmacy; Shanghai Jiao Tong University; Shanghai, PR China
| |
Collapse
|
29
|
T lymphocytes from chronic HCV-infected patients are primed for activation-induced apoptosis and express unique pro-apoptotic gene signature. PLoS One 2013; 8:e77008. [PMID: 24130824 PMCID: PMC3794995 DOI: 10.1371/journal.pone.0077008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/28/2013] [Indexed: 12/12/2022] Open
Abstract
Although extensive studies have demonstrated the functional impairment of antigen-specific CD4(+) and CD8(+) T-cells during chronic hepatitis C virus (HCV) infection, the functional status of global CD4(+) and CD8(+) T-cells remains unclear. In this report, we recruited 42 long-term (~20 years) treatment-naïve chronic HCV (CHC) patients and 15 healthy donors (HDs) to investigate differences in global CD4(+) and CD8(+) T-cells function. We show that CD4(+) and CD8(+) T-cells from CHC patients underwent increased apoptosis after TCR stimulation. Furthermore, IFN-γ, IL-9 and IP-10 were elevated in CHC patients' plasma and promoted activation-induced T-cells death. Global CD4(+) and CD8(+) T-cells also showed unique transcriptional profiles in the expression of apoptosis-related genes. We identified BCL2, PMAIP1, and CASP1 in CD4(+) T-cells and IER3 and BCL2A1 in CD8(+) T-cells from CHC patients as HCV-specific gene signatures. Importantly, the gene expression patterns of CD4(+) and CD8(+) T-cells from CHC patients differ from those in CD4(+) and CD8(+) T-cells from human immunodeficiency virus type 1 (HIV-1) or hepatitis B virus (HBV) infected individuals. Our results indicate that chronic HCV infection causes a systemic change in cytokine levels that primes T-cells for activation-induced apoptosis. Furthermore, HCV infection programs unique apoptosis-related gene expression profiles in CD4(+) and CD8(+) T-cells, leading to their enhanced activation-induced apoptosis. These results provide novel insights to the pathogenesis of chronic HCV infection.
Collapse
|
30
|
Fishbourne E, Hutet E, Abrams C, Cariolet R, Le Potier MF, Takamatsu HH, Dixon LK. Increase in chemokines CXCL10 and CCL2 in blood from pigs infected with high compared to low virulence African swine fever virus isolates. Vet Res 2013; 44:87. [PMID: 24083897 PMCID: PMC3832245 DOI: 10.1186/1297-9716-44-87] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 09/05/2013] [Indexed: 11/17/2022] Open
Abstract
Modulation of the expression of chemokines and chemokine receptors in whole blood was compared following infection of pigs with high and low virulence isolates of African swine fever virus. Levels of mRNAs for CCL2, CCL3L1, CCL4, CXCL10, CCR1 and CCR5 were significantly increased in at least one time point following infection in two experiments and CCL5, CCR9 and CXCR4 mRNA were significantly increased in one of the experiments. The results showed that greatest fold increases in mRNAs for CXCL10 and CCL2 were observed following infection of pigs. CXCL10 mRNA was increased by up to 15 fold in infected compared to uninfected pigs. CXCL10 protein was also detected in serum from pigs infected with the high virulence Benin 97/1 isolate. Levels of CCL2 mRNA were increased in pigs infected with high virulence Benin 97/1 isolate compared to low virulence OURT88/3 isolate and this correlated with an increase of greater than 30 fold in levels of CCL2 protein detected in serum from pigs infected with this isolate. An increase in overall chemotaxis active compounds in defibrinated plasma samples from Benin 97/1 infected pigs was observed at 3 days post-infection (dpi) and a decrease by 7 dpi as measured by chemotaxis assay using normal pig leucocytes in vitro. Increased levels of CXCL10 may either contribute to the activation of lymphocyte priming toward the Th1 phenotype or induction of T lymphocyte apoptosis. Increased levels of CCL2, a chemoattractant for macrophages, may result in increased recruitment of monocytes from bone marrow thus increasing the pool of cells susceptible to infection.
Collapse
Affiliation(s)
- Emma Fishbourne
- The Pirbright Institute, Pirbright, Woking, Surrey GU24 0NF, UK.
| | | | | | | | | | | | | |
Collapse
|
31
|
Gene Expression Analysis of the IPEC-J2 Cell Line: A Simple Model for the Inflammation-Sensitive Preterm Intestine. ACTA ACUST UNITED AC 2013. [DOI: 10.1155/2013/980651] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The IPEC-J2 cell line was studied as a simple model for investigating responses of the newborn intestinal epithelium to diets. Especially, the small intestine of immature newborns is sensitive to diet-induced inflammation. We investigated gene expression of epithelial- and immune response-related genes in IPEC-J2 cells stimulated for 2 h with milk formula (CELL-FORM), colostrum (CELL-COLOS), or growth medium (CELL-CONTR) and in distal small intestinal tissue samples from preterm pigs fed milk formula (PIG-FORM) or colostrum (PIG-COLOS). High throughput quantitative PCR analysis of 48 genes revealed the expression of 22 genes in IPEC-J2 cells and 31 genes in intestinal samples. Principal component analysis (PCA) discriminated the gene expression profile of IPEC-J2 cells from that of intestinal samples. The expression profile of intestinal tissue was separated by PCA into 2 groups according to diet, whereas no diet-dependent grouping was seen for IPEC-J2 cells. Expression differences between PIG-FORM and PIG-COLOS were found for DEFB1, CXCL10, IL1RN, and ALPI, while IL8 was upregulated in CELL-FORM compared with CELL-CONTR. These differences, between IPEC-J2 cells and intestinal tissue from preterm pigs, both used as models for the newborn intestine, underline that caution must be exercised prior to analysis and interpretation of diet-induced effects on gene expression.
Collapse
|
32
|
Lai W, Wu J, Zou X, Xie J, Zhang L, Zhao X, Zhao M, Wang Q, Ji J. Secretome Analyses of Aβ1–42 Stimulated Hippocampal Astrocytes Reveal that CXCL10 is Involved in Astrocyte Migration. J Proteome Res 2013; 12:832-43. [DOI: 10.1021/pr300895r] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Wenjia Lai
- The National Laboratory of Protein
Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, P. R. China
| | - Jing Wu
- The National Laboratory of Protein
Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, P. R. China
| | - Xiao Zou
- The National Laboratory of Protein
Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, P. R. China
| | - Jian Xie
- Department of Neurosurgery, Beijing TianTan Hospital Affiliated to Capital Medical University, Beijing 100875, P. R. China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing TianTan Hospital Affiliated to Capital Medical University, Beijing 100875, P. R. China
| | - Xuyang Zhao
- The National Laboratory of Protein
Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, P. R. China
| | - Minzhi Zhao
- The National Laboratory of Protein
Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, P. R. China
| | - Qingsong Wang
- The National Laboratory of Protein
Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, P. R. China
| | - Jianguo Ji
- The National Laboratory of Protein
Engineering and Plant Genetic Engineering, College of Life Sciences, Peking University, Beijing 100871, P. R. China
| |
Collapse
|