1
|
Dai Y, Wu J, Wang J, Wang H, Guo B, Jiang T, Cai Z, Han J, Zhang H, Xu B, Zhou X, Wang C. Magnesium Ions Promote the Induction of Immunosuppressive Bone Microenvironment and Bone Repair through HIF-1α-TGF-β Axis in Dendritic Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311344. [PMID: 38661278 DOI: 10.1002/smll.202311344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 03/22/2024] [Indexed: 04/26/2024]
Abstract
The effect of immunoinflammation on bone repair during the recovery process of bone defects needs to be further explored. It is reported that Mg2+ can promote bone repair with immunoregulatory effect, but the underlying mechanism on adaptive immunity is still unclear. Here, by using chitosan and hyaluronic acid-coated Mg2+ (CSHA-Mg) in bone-deficient mice, it is shown that Mg2+ can inhibit the activation of CD4+ T cells and increase regulatory T cell formation by inducing immunosuppressive dendritic cells (imDCs). Mechanistically, Mg2+ initiates the activation of the MAPK signaling pathway through TRPM7 channels on DCs. This process subsequently induces the downstream HIF-1α expression, a transcription factor that amplifies TGF-β production and inhibits the effective T cell function. In vivo, knock-out of HIF-1α in DCs or using a HIF-1α inhibitor PX-478 reverses inhibition of bone inflammation and repair promotion upon Mg2+-treatment. Moreover, roxadustat, which stabilizes HIF-1α protein expression, can significantly promote immunosuppression and bone repair in synergism with CSHA-Mg. Thus, the findings identify a key mechanism for DCs and its HIF-1α-TGF-β axis in the induction of immunosuppressive bone microenvironment, providing potential targets for bone regeneration.
Collapse
Affiliation(s)
- Yuya Dai
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Jinhui Wu
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Junyou Wang
- State-Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Haoze Wang
- Nation Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Bingqing Guo
- Changzhou Hospital of Traditional Chinese Medicine, Changzhou, 213000, China
| | - Tao Jiang
- Changzhou Hospital of Traditional Chinese Medicine, Changzhou, 213000, China
| | - Zhuyun Cai
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Junjie Han
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Haoyu Zhang
- Nation Key Laboratory of Medical Immunology, Institute of Immunology, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Bangzhe Xu
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Xuhui Zhou
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Ce Wang
- Department of Orthopedic, Changzheng Hospital Affiliated to Naval Medical University, Shanghai, 200003, China
| |
Collapse
|
2
|
Sánchez-Ponce Y, Murillo-Eliosa JR, Morales-Sanchez A, Fuentes-Pananá EM. Cytokine Dynamics and Herpesvirus Interactions in Pediatric Liver and Kidney Transplant Recipients: The Distinct Behavior of HCMV, HHV6, HHV7 and EBV. Viruses 2024; 16:1067. [PMID: 39066229 PMCID: PMC11281388 DOI: 10.3390/v16071067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Pediatric solid organ transplant (SOT) recipients face a challenging balance between immunosuppression and graft rejection. While Epstein-Barr Virus (EBV) and cytomegalovirus (HCMV) are known contributors to post-transplant lymphoproliferative disease and graft rejection, respectively, the roles of herpesvirus 6 and 7 (HHV6 and HHV7) and the impact of these herpesviruses on cytokine levels remain unclear, leading to gaps in clinical practice. In this associative study, we measured 17 cytokines using a Bio-Plex assay in a meticulously curated plasma sample pool (N = 158) from pediatric kidney and liver transplant recipients over a one-year follow-up period. The samples included virus-negative and virus-positive cases, either individually or in combination, along with episodes of graft rejection. We observed that the elevation of IL-4, IL-8, and IL-10 correlated with graft rejection. These cytokines were elevated in samples where HCMV or HHV6 were detected alone or where EBV and HHV7 were co-detected. Interestingly, latent EBV, when detected independently, exhibited an immunomodulatory effect by downregulating cytokine levels. However, in co-detection scenarios with β-herpesviruses, EBV transitioned to a lytic state, also associating with heightened cytokinemia and graft rejection. These findings highlight the complex interactions between the immune response and herpesviruses in transplant recipients. The study advocates for enhanced monitoring of not only EBV and HCMV but also HHV6 and HHV7, providing valuable insights for improved risk assessment and targeted interventions in pediatric SOT recipients.
Collapse
Affiliation(s)
- Yessica Sánchez-Ponce
- Research Unit in Virology and Cancer, Children’s Hospital of Mexico Federico Gómez, Mexico City 06720, Mexico; (Y.S.-P.); (A.M.-S.)
- Postgraduate Program in Biological Science, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | | | - Abigail Morales-Sanchez
- Research Unit in Virology and Cancer, Children’s Hospital of Mexico Federico Gómez, Mexico City 06720, Mexico; (Y.S.-P.); (A.M.-S.)
| | | |
Collapse
|
3
|
Wang X, Mo X, Yang Z, Zhao C. Qntrolling the LncRNA HULC-Tregs-PD-1 axis inhibits immune escape in the tumor microenvironment. Heliyon 2024; 10:e28386. [PMID: 38560250 PMCID: PMC10979100 DOI: 10.1016/j.heliyon.2024.e28386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Background Immune escape remains a major challenge in the treatment of malignant tumors. Here, we studied the mechanisms underlying immune escape in the tumor microenvironment and identified a potential therapeutic target. Methods Pathological specimens from patients with liver cancer, soft tissue sarcoma, and liver metastasis of colon cancer were subjected to immunohistochemistry analysis to detect the expression of programmed death-1 (PD-1) in the tumor microenvironment (TME). Additionally, the expression of regulatory T cells (Tregs) and long non-coding RNAs (lncRNAs), such as highly upregulated in liver cancer (HULC) was evaluated by fluorescence in situ hybridization, and the relationship between HULC, Treg cells, and PD-1 was determined. The animals were divided into H22 hepatic carcinoma and S180 sarcoma groups. Each group was divided into Foxp3-/-C57BL/6J and C57BL/6J mice. Thereafter, mice were inoculated with 0.1 ml S180 sarcoma cells or 0.1 ml H22 hepatoma cells, at a concentration of 1 × 107/ml. The number of splenic CD4+CD25+Foxp3+ T cells was detected by flow cytometry, and serum interleukin-10 (IL-10) and transforming growth factor β1 (TGF-β1) levels were detected using a Luminex liquid suspension chip. Expression of PD-1, fork head box P3 (Foxp3), and HULC in the TME, were analyzed and the therapeutic effect of inhibiting the lncRNA HULC-Treg-PD-1 axis in malignant tumors was determined. Results High expression of lncRNA HULC promotes the proliferation of Treg cells and increases PD-1 expression in the tumor microenvironment. The HULC-Treg-PD-1 axis plays an immunosuppressive role and promotes the proliferation of malignant tumors. Knocking out the Foxp3 gene can affect the HULC-Treg-PD-1 axis and reduce PD-1, IL-10, and TGF-β1 expression to control the growth of malignant tumors. Conclusion The lncRNA HULC-Treg-PD-1 axis promotes the growth of malignant tumors. This axis could be modulated to reduce PD-1, IL-10, and TGF-β1 expression and the subsequent immune escape. The inhibition of immune escape in the tumor microenvironment can be achieved by controlling the LncRNA HULC-Treg-PD-1 axis.
Collapse
Affiliation(s)
- XiaoYu Wang
- School of Health Science, Guangdong Pharmaceutical University, Guangzhou, 51006, China
| | - Xiaoyan Mo
- The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, 51006, China
| | - Zhuolin Yang
- School of Health Science, Guangdong Pharmaceutical University, Guangzhou, 51006, China
| | - Changlin Zhao
- School of Health Science, Guangdong Pharmaceutical University, Guangzhou, 51006, China
- The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, 51006, China
| |
Collapse
|
4
|
Wang J, Ma Y, Wang J. miR-27a-5p inhibits acute rejection of liver transplantation in rats by inducing M2 polarization of Kupffer cells through the PI3K/Akt pathway. Cytokine 2023; 165:156085. [PMID: 37003239 DOI: 10.1016/j.cyto.2022.156085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/28/2022] [Accepted: 11/01/2022] [Indexed: 04/03/2023]
Abstract
Liver transplantation (LT), a major therapy for end-stage liver disease, is often associated with acute rejection (AR). MicroRNAs (miRNAs) have been implicated in AR-related gene regulation. In this experiment, the mechanism of miR-27a-5p in AR of LT was studied. Allotransplantation model (LEW-BN) and syngeneic transplantation model (LEW-LEW) of rat orthotopic liver transplantation (OLT) were established. miR-27a-5p was overexpressed in recipient rats 28 days before LT to detect its effects on LT pathology, liver function, and survival time. Kupffer cells (KCs) were isolated and treated with lipopolysaccharide (LPS) and miR-27a-5p overexpression. miR-27a-5p overexpression reduced lymphocyte numbers around portal areas and central veins after LT and mitigated degeneration of epithelial cells of the bile duct. Expression levels of IL-10 and TGF-β1 were increased while IL-12 was decreased. Liver function damage was alleviated and the survival time of rats with LT was prolonged. miR-27a-5p induced M2 polarization of rats with AR after LT and LPS-treated KCs in vitro and promoted activation of the PI3K/Akt pathway in KCs. Inhibition of the PI3K/Akt pathway averted induction of miR-27a-5p on M2 polarization of KCs. Taken together, miR-27a-5p inhibited AR after LT in rats by inducing M2 polarization of KCs through the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Jian Wang
- School of Physical Education Shanxi University, 030006 Taiyuan, China
| | - Yuanyuan Ma
- Research Center for Health Promotion of Children and Adolescents, Taiyuan Institute of Technology, No. 31, Xinlan Road, Jiancaoping District, 030008 Taiyuan, China.
| | - Jinxian Wang
- Research Center for Health Promotion of Children and Adolescents, Taiyuan Institute of Technology, No. 31, Xinlan Road, Jiancaoping District, 030008 Taiyuan, China
| |
Collapse
|
5
|
Cao W, Lu J, Li S, Song F, Xu J. Transcriptomic analysis of graft liver provides insight into the immune response of rat liver transplantation. Front Immunol 2022; 13:947437. [DOI: 10.3389/fimmu.2022.947437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/07/2022] [Indexed: 11/05/2022] Open
Abstract
BackgroundAs an “immune-privileged organ”, the liver has higher rates of both spontaneous tolerance and operational tolerance after being transplanted compared with other solid organs. Also, a large number of patients still need to take long-term immunosuppression regimens. Liver transplantation (LT) rejection involves varieties of pathophysiological processes and cell types, and a deeper understanding of LT immune response is urgently needed.MethodsHomogenic and allogeneic rat LT models were established, and recipient tissue was collected on postoperative day 7. The degree of LT rejection was evaluated by liver pathological changes and liver function. Differentially expressed genes (DEGs) were detected by transcriptome sequencing and confirmed by reverse transcription-polymerase chain reaction. The functional properties of DEGs were characterized by the Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome pathway analyses. The cells infiltrating the graft and recipient spleen and peripheral blood were evaluated by immunofluorescence and flow cytometry.ResultA total of 1,465 DEGs were screened, including 1,177 up-regulated genes and 288 down-regulated genes. GO enrichment and KEGG pathway analysis indicated that DEGs were involved in several immunobiological processes, including T cell activation, Th1, Th2 and Th17 cell differentiation, cytokine-cytokine receptor interaction and other immune processes. Reactome results showed that PD-1 signaling was enriched. Further research confirmed that mRNA expression of multiple immune cell markers increased and markers of T cell exhaustion significantly changed. Flow cytometry showed that the proportion of Treg decreased, and that of PD-1+CD4+ T cells and PD-1+CD8+ T cells increased in the allogeneic group.ConclusionUsing an omic approach, we revealed that the development of LT rejection involved multiple immune cells, activation of various immune pathways, and specific alterations of immune checkpoints, which would benefit risk assessment in the clinic and understanding of pathogenesis regarding LT tolerance. Further clinical validations are warranted for our findings.
Collapse
|
6
|
Gama JFG, Cardoso LMDF, Bisaggio RDC, Lagrota-Candido J, Henriques-Pons A, Alves LA. Immunological Tolerance in Liver Transplant Recipients: Putative Involvement of Neuroendocrine-Immune Interactions. Cells 2022; 11:cells11152327. [PMID: 35954171 PMCID: PMC9367574 DOI: 10.3390/cells11152327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/20/2022] [Accepted: 06/29/2022] [Indexed: 02/04/2023] Open
Abstract
The transplantation world changed significantly following the introduction of immunosuppressants, with millions of people saved. Several physicians have noted that liver recipients that do not take their medication for different reasons became tolerant regarding kidney, heart, and lung transplantations at higher frequencies. Most studies have attempted to explain this phenomenon through unique immunological mechanisms and the fact that the hepatic environment is continuously exposed to high levels of pathogen-associated molecular patterns (PAMPs) or non-pathogenic microorganism-associated molecular patterns (MAMPs) from commensal flora. These components are highly inflammatory in the periphery but tolerated in the liver as part of the normal components that arrive via the hepatic portal vein. These immunological mechanisms are discussed herein based on current evidence, although we hypothesize the participation of neuroendocrine-immune pathways, which have played a relevant role in autoimmune diseases. Cells found in the liver present receptors for several cytokines, hormones, peptides, and neurotransmitters that would allow for system crosstalk. Furthermore, the liver is innervated by the autonomic system and may, thus, be influenced by the parasympathetic and sympathetic systems. This review therefore seeks to discuss classical immunological hepatic tolerance mechanisms and hypothesizes the possible participation of the neuroendocrine-immune system based on the current literature.
Collapse
Affiliation(s)
- Jaciara Fernanda Gomes Gama
- Laboratory of Cellular Communication, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Brazil Avenue, 4365-Manguinhos, Rio de Janeiro 21045-900, Brazil; (J.F.G.G.); (L.M.d.F.C.)
- Laboratory of Immunopathology, Department of Immunobiology, Biology Institute, Federal Fluminense University (UFF), Gragoatá Bl-M Campus, Niterói 24210-200, Brazil;
| | - Liana Monteiro da Fonseca Cardoso
- Laboratory of Cellular Communication, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Brazil Avenue, 4365-Manguinhos, Rio de Janeiro 21045-900, Brazil; (J.F.G.G.); (L.M.d.F.C.)
| | - Rodrigo da Cunha Bisaggio
- Department of Biotechnology, Federal Institute of Rio de Janeiro (IFRJ), Maracanã, Rio de Janeiro 20270-021, Brazil;
| | - Jussara Lagrota-Candido
- Laboratory of Immunopathology, Department of Immunobiology, Biology Institute, Federal Fluminense University (UFF), Gragoatá Bl-M Campus, Niterói 24210-200, Brazil;
| | - Andrea Henriques-Pons
- Laboratory of Innovations in Therapies, Education, and Bioproducts, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-361, Brazil;
| | - Luiz A. Alves
- Laboratory of Cellular Communication, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Brazil Avenue, 4365-Manguinhos, Rio de Janeiro 21045-900, Brazil; (J.F.G.G.); (L.M.d.F.C.)
- Correspondence: or ; Tel.: +55-(21)-2562-1816 (ext. 1841)
| |
Collapse
|
7
|
Assadiasl S, Mooney N, Nicknam MH. Cytokines in Liver Transplantation. Cytokine 2021; 148:155705. [PMID: 34564024 DOI: 10.1016/j.cyto.2021.155705] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 08/17/2021] [Accepted: 09/10/2021] [Indexed: 02/08/2023]
Abstract
Cytokines, soluble mediators of the immune system, play a critical role in the pathogenesis of autoimmune, allergic and infectious diseases. They are also implicated in the initiation and development of allograft rejection. During recent years, there have been considerable advances in generating novel anti-cytokine agents with promoted efficacy and safety, which could be administrated for managing dysregulated cytokine secretion; besides, gene therapy for overexpression of immunomodulatory cytokines has shown substantial improvements. Liver transplantation has been established as a life-saving treatment for end-stage hepatic diseases but the growing number of recipients urge for improved post-transplant care including tolerance induction, infection control and resolving immunosuppressant drugs adverse effects. Cytokines with a wide range of proinflammatory and regulatory properties might be considered as potential therapeutic targets for selective suppression or enhancement of the immune responses in recipients. In the present review, we aimed to summarize the positive and negative effects of cytokines on liver allograft in addition to their prognostic and therapeutic values.
Collapse
Affiliation(s)
- Sara Assadiasl
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nuala Mooney
- Human Immunology and Immunopathology, Inserm UMR 976, Paris, France; Université de Paris, Paris, France
| | - Mohammad Hossein Nicknam
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, Medical School, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Xiong A, Wang J, Mao XL, Jiang Y, Fan Y. MiR‐199a‐3p modulates the function of dendritic cells involved in transplantation tolerance by targeting CD86. HLA 2019; 94:493-503. [PMID: 31448543 DOI: 10.1111/tan.13677] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 08/06/2019] [Accepted: 08/23/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Ali Xiong
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| | - Jing Wang
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| | - Xiao Li Mao
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| | - Yi Jiang
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| | - Yue Fan
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| |
Collapse
|
9
|
Chen L, Zhang L, Zhu Z, He W, Gao L, Zhang W, Liu J, Huang A. Effects of IL-10- and FasL-overexpressing dendritic cells on liver transplantation tolerance in a heterotopic liver transplantation rat model. Immunol Cell Biol 2019; 97:714-725. [PMID: 30977930 DOI: 10.1111/imcb.12252] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 12/14/2022]
Abstract
Acute rejection is the major determinant for the long-term survival of donor liver after liver transplantation (LT). The aim of this study was to examine the therapeutic potential of interleukin (IL)-10-FasL-overexpressing immature dendritic cells (imDCs) to induce local immunosuppression in liver grafts. imDCs derived from donors were transduced by lentiviral vectors expressing human IL-10 and/or Fas ligand (FasL) gene(s), and the expression of surface molecules and the ability to induce T-cell proliferation were measured. imDCs were intraperitoneally injected into recipient rats as a model of LT to examine the rejection grade [Banff rejection activity index (RAI)], liver functions [Alanine aminotransferase, Aspartate aminotransferase (AST) and total bilirubin (TBIL)] and post-transplant survival. IL-10 and FasL co-transduction of imDCs induced a greater reduction in CD80, CD86 and major histocompatibility complex class II (MHC II) expression, as well as T-cell proliferation, but increased levels of IL-10 and FasL in culture supernatants compared with mono-transduced or untransduced imDCs (P < 0.05). The infusion of co-transduced imDCs in LT recipients reduced RAI scores, decreased plasma AST and TBIL, and prolonged survival compared with mono-transduced or untransduced imDC-treated liver allografts. These findings demonstrated that the transfusion of IL-10-FasL/imDCs enhanced immune tolerance and prolonged the survival of liver allografts after LT. The immunomodulatory activity of IL-10- and FasL-modified imDCs might be a new therapeutic approach to prevent organ rejection in clinical transplantation.
Collapse
Affiliation(s)
- Lihong Chen
- Department of Pathology, School of Basic Medical Sciences of Fujian Medical University, Fuzhou, Fujian, China.,Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China.,Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, China.,Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Lina Zhang
- Department of Pathology, School of Basic Medical Sciences of Fujian Medical University, Fuzhou, Fujian, China.,Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Zhu Zhu
- Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Wubing He
- Provincial Clinical Medical College, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Lingyun Gao
- Department of Pathology, School of Basic Medical Sciences of Fujian Medical University, Fuzhou, Fujian, China.,Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China.,Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, China.,Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenmin Zhang
- Department of Pathology, School of Basic Medical Sciences of Fujian Medical University, Fuzhou, Fujian, China.,Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, China.,Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Jingfeng Liu
- Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China.,Divison of Hepatobiliary Surgery, Hepatic Disease Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Aimin Huang
- Department of Pathology, School of Basic Medical Sciences of Fujian Medical University, Fuzhou, Fujian, China.,Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, China.,Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
10
|
Lyu C, Qu Y, Mu DZ. [Research advances in immune tolerance of allogeneic cell transplantation in preterm infants]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:338-340. [PMID: 29658462 PMCID: PMC7390031 DOI: 10.7499/j.issn.1008-8830.2018.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/10/2018] [Indexed: 06/08/2023]
Abstract
Preterm infants are a special group, and related severe neurological, respiratory, and digestive disorders have high disability/fatality rates. Allogeneic cell transplantation may be an effective method for the prevention and treatment of these diseases. At present, animal studies have been conducted for allogeneic cell transplantation in the treatment of hypoxic-ischemic encephalopathy, bronchopulmonary dysplasia, and necrotizing enterocolitis. The main difficulty of this technique is graft-versus-host reaction (GVHR), and successful induction of immune tolerance needs to be achieved in order to solve this problem. This article reviews the research advances in immune tolerance of allogeneic cell transplantation in preterm infants.
Collapse
Affiliation(s)
- Can Lyu
- Department of Pediatrics, West China Second University Hospital/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China.
| | | | | |
Collapse
|
11
|
Huang C, Zhang L, Ling F, Wen S, Luo Y, Liu H, Liu J, Zheng W, Liang M, Sun J, Lin YK. Effect of immune tolerance induced by immature dendritic cells and CTLA4-Ig on systemic lupus erythematosus: An in vivo study. Exp Ther Med 2018; 15:2499-2506. [PMID: 29456655 DOI: 10.3892/etm.2018.5697] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 07/27/2017] [Indexed: 11/06/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multi-organ autoimmune disease in which tissue damage is caused by autoantibodies. The induction of specific immune tolerance, including the utilization of immune regulatory cells, may enhance the therapeutic effects of organ transplantation in patients with SLE. Furthermore, inhibiting immune responses has been reported to be an effective treatment for SLE. However, few studies have explored the association between an increased immune tolerance and a decreased immune response in SLE treatment. Dendritic cells (DCs), which are highly efficient antigen-presenting cells, are able to induce specific tolerance, while cytotoxic T lymphocyte antigen 4-immunoglobulin (CTLA4-Ig) inhibits the immune response. In the present study, interleukin (IL)-10-treated DCs and CTLA4-Ig were administered to mice with SLE alone or in combination and the therapeutic effects were investigated. IL-10 was added into the culture medium of bone marrow-derived DCs to prevent them from differentiating into mature cells. Low levels of major histocompatibility complex II, cluster of differentiation (CD)40, CD80 and CD86 were detected, which indicated that the immature state of DCs was maintained. IL-10-treated DCs were subsequently injected into the caudal vein of B6.MRL-Faslpr/J lupus mice, which are an established animal model of SLE. To amplify the tolerance effect, mice were simultaneously injected with CTLA4-Ig. Compared with the IL-10-treated DC and CTLA4-Ig groups, combined treatment with IL-10-treated DCs and CTLA4-Ig strongly induced immune tolerance in mice with SLE, as indicated by the significantly reduced levels of urine protein, anti-nuclear antibody, double-stranded DNA and IL-17A. A significant decrease in the proportion of T helper cells and an increase in the proportion of CD4+ forkhead box protein P3+ Treg cells was also observed, further confirming the induction of immune tolerance. These results suggest that combined treatment with IL-10-DCs and CTLA4-Ig may be a promising novel therapeutic strategy for the treatment of SLE.
Collapse
Affiliation(s)
- Cuili Huang
- Department of Dermatology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Lidan Zhang
- Department of Dermatology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Fang Ling
- Department of Cell Biology and Genetics, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Sijian Wen
- Department of Dermatology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yanyan Luo
- Department of Dermatology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Hui Liu
- Department of Dermatology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jingping Liu
- Department of Dermatology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Wenjun Zheng
- Department of Dermatology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Ming Liang
- Department of Dermatology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jian Sun
- Department of Cell Biology and Genetics, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - You-Kun Lin
- Department of Dermatology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
12
|
Li RD, Tao YF, Shen CH, Ma ZY, Zhang XF, Zhang QB, Wang ZX. Role of Th17 and Treg cells and Th17/Treg imbalance in immune tolerance in rats after liver transplantation. Shijie Huaren Xiaohua Zazhi 2017; 25:3046-3052. [DOI: 10.11569/wcjd.v25.i34.3046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the relationship between Th17/Treg expression in liver tissue and immune tolerance after liver transplantation.
METHODS A rat model of orthotopic liver transplantation from Lewis rats to Brown Norway (BN) rats (LEWIS→BN) was developed using Kamada's two-cuff technique. Control rats underwent orthotopic BN→BN liver transplantation. After operation, the general condition of rats was observed. Rats were sacrificed on days 1, 3, 5, and 7 after modeling to collect liver tissues for HE staining. The levels of serum alanine transaminase (ALT), aspartate transaminase (AST), interleukin (IL)-17, IL-23, IL-10, and transforming growth factor (TGF)-β1 were measured by ELISA.
RESULTS Compared with the control group, serum levels of ALT and AST were increased obviously in the LEWIS→BN group at all the three time points (P < 0.05, on the 7th day, ALT in the control group: 819.29 IU/L ± 79.33 IU/L; in the LEWIS→BN group: 1305.62 IU/L ± 94.82 IU/L, AST in the control group: 337.82 IU/L ± 32.17 IU/L; AST in the LEWIS→BN group: 867.75 IU/L ± 73.97 IU/L); the levels of Th17 related factors IL-17 (the control group: 28.67 pg/mL ± 2.55 pg/mL, the LEWIS→BN group: 92.36 pg/mL ± 9.00 pg/mL) and IL-23 (the control group: 26.82 pg/mL ± 8.17 pg/mL, the LEWIS→BN group: 62.98 pg/mL ± 12.96 pg/mL) in the peripheral blood were increased dramatically, while the levels of Treg related factor IL-10 (the control group: 76.92 pg/mL ± 12.87 pg/mL, the LEWIS→BN group: 47.92 pg/mL ± 7.00 pg/mL) and TGF-β1 (the control group: 129.47 pg/mL ± 18.37 pg/mL, the LEWIS→BN group: 82.48 pg/mL ± 11.83 pg/mL) were decreased dramatically in the LEWIS→BN group, suggesting the presence of Th17/Treg imbalance in acute rejection. The downregulation of Smad2/3 protein levels had the same trend as TGF-β1, and the difference between the Lewis→BN acute rejection group and the BN→ BN immune tolerance group was statistically significant (P < 0.05).
CONCLUSION IL-17, IL-23, IL-10, and TGF-β1 take part in acute rejection after orthotopic liver transplantation, causing Treg to Th17 immune deviation, which may be used as a target index in immune tolerance of liver transplantation.
Collapse
Affiliation(s)
- Rui-Dong Li
- Department of Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Yi-Feng Tao
- Department of Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Cong-Huan Shen
- Department of Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Zhen-Yu Ma
- Department of Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Xiao-Fei Zhang
- Department of Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Quan-Bao Zhang
- Department of Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Zheng-Xin Wang
- Department of Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, China
| |
Collapse
|
13
|
Qiu M, Chen Y, Chen L, Zeng J, Liu J. Transforming growth factor β1 and Fas ligand synergistically enhance immune tolerance in dendritic cells in liver transplantation. J Surg Res 2017; 218:180-193. [PMID: 28985848 DOI: 10.1016/j.jss.2017.05.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/08/2017] [Accepted: 05/11/2017] [Indexed: 01/03/2023]
Affiliation(s)
- Minglian Qiu
- Department of Thoracic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China.
| | - Yujuan Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Lihong Chen
- Department of Pathology, School of Basic Medical Sciences of Fujian Medical University, Fuzhou, Fujian, China
| | - Jinhua Zeng
- Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jingfeng Liu
- Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
14
|
He W, Chen L, Zheng L, Luo L, Gao L. Prolonged survival effects induced by immature dendritic cells and regulatory T cells in a rat liver transplantation model. Mol Immunol 2016; 79:92-97. [PMID: 27764710 DOI: 10.1016/j.molimm.2016.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/09/2016] [Accepted: 10/12/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Dendritic cells (DCs) and regulatory T (Treg) cells are crucial for inducing immune tolerance. However, the suppressive function of infused Treg cells and immature DCs (imDCs) following solid organ transplantation remains unclear. METHODS ImDCs derived from DA-donor rats and Treg cells isolated from spleens of Lewis rats were prepared. A heterotopic liver transplantation model was established to examine the immune tolerance effects of infusion of Treg-imDCs, imDCs and Treg cells individually. Th1/Th2 cytokines and TRAL were detected by ELISA. The overall rejection grade was assessed and the rejection activity index (RAI) was calculated. TUNEL-positive lymphocytes were detected in the portal area in liver sections. RESULTS The infusion of Treg-imDCs was more effective than imDCs or Treg cells individually. Moreover, the expression of IL-10 and TGF-β1 was significantly up-regulated, and IL-12 expression was significantly down-regulated, especially in the Treg-imDCs group. The percentage of TUNEL-positive cells was significantly higher in the Treg cells and imDCs groups. The RAI values in Treg-imDCs group on days 3 and 7 were lower than control, imDCs and Treg cells groups individually (p<0.05). Both TBIL and ALT levels in the Treg-imDCs and imDCs groups were significantly lower than those of the control and Treg cells groups, and serum TRAL levels increased significantly 10days after transplantation in the imDC and Treg-imDC groups compared with the control and Treg cells groups (P<0.001). CONCLUSION These data demonstrated that infusion of Treg cells and/or imDCs induces alloantigen tolerance and prolongs liver allograft survival. The infusion of Treg-imDCs was more effective than imDCs or Treg cells individually. ImDCs synergize with Treg cells in inducing and maintaining the feedback loop between imDCs and Treg cells in vivo.
Collapse
Affiliation(s)
- Wubing He
- Fujian Provincial Hospital, Provincial Clinical Medical College, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Lihong Chen
- Department of Pathology, School of Basic Medical Sciences of Fujian Medical University, Fuzhou, Fujian 350004, China; Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350025, China.
| | - Lin Zheng
- Department of Pathology, School of Basic Medical Sciences of Fujian Medical University, Fuzhou, Fujian 350004, China
| | - Liuping Luo
- Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian 350025, China
| | - Lingyun Gao
- Department of Pathology, School of Basic Medical Sciences of Fujian Medical University, Fuzhou, Fujian 350004, China
| |
Collapse
|
15
|
Aurelian L, Bollino D, Colunga A. The oncolytic virus ΔPK has multimodal anti-tumor activity. Pathog Dis 2016; 74:ftw050. [PMID: 27242376 DOI: 10.1093/femspd/ftw050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2016] [Indexed: 02/07/2023] Open
Abstract
Oncolytic viruses (OVs) are an emerging cancer therapeutic, with a near complete absence of serious adverse effects. However, clinical efficacy is relatively modest, related to poor tumor penetration, failure to lyse cancer stem cells (CSCs) and blockade of immunogenic cell death by the immunosuppressive tumor microenvironment. To overcome such limitations, we developed an OV (known as ΔPK) with multimodal anti-tumor activity. ΔPK has potent anti-tumor activity both in melanoma cell lines and xenograft animal models, associated with virus replication and the induction of multiple independent programmed cell death pathways. It lyses CSCs through autophagy modulation and it reverses the immunosuppressive tumor microenvironment by altering the balance of cytokines secreted by the tumor cells. This includes decreased tumor cell secretion of the immunosuppressive and procancerous cytokines IL-10 and IL-18 and concomitant increased secretion of the proinflammatory cytokines TNF-α, GM-CSF, IL-6 and IL-1β. ΔPK also upregulates the NKG2D ligand, MICA expressed by cytotoxic NK and T cells, and downregulates the negative immune checkpoint regulator cytotoxic T-lymphocyte antigen-4 (CTLA-4). ΔPK is well tolerated in human patients in whom it also alters the Th1/Th2 balance. Further studies are designed to elucidate the role of these contributions in different tumor types.
Collapse
Affiliation(s)
- Laure Aurelian
- Department of Microbiology and Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dominique Bollino
- Department of Microbiology and Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Aric Colunga
- Department of Microbiology and Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
16
|
Bollino D, Colunga A, Li B, Aurelian L. ΔPK oncolytic activity includes modulation of the tumour cell milieu. J Gen Virol 2015; 97:496-508. [PMID: 26602205 DOI: 10.1099/jgv.0.000353] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Oncolytic virotherapy is a unique cancer therapeutic that encompasses tumour cell lysis through both virus replication and programmed cell death (PCD) pathways. Nonetheless, clinical efficacy is relatively modest, likely related to the immunosuppressive tumour milieu. Our studies use the herpes simplex virus type 2 (HSV-2)-based oncolytic virus ΔPK that has documented anti-tumour activity associated with virus replication, PCD and cancer stem cell lysis. They are designed to examine whether ΔPK-mediated oncolysis includes the ability to reverse the immunosuppressive tumour microenvironment by altering the balance of cytokines directly secreted by the melanoma cells and to define its mechanism. Here, we show that melanoma cells secreted the immunosuppressive cytokine IL-10, and that secretion was inhibited by ΔPK through virus replication and c-Jun N-terminal kinase/c-Jun activation. ΔPK-induced IL-10 inhibition upregulated surface expression of MHC class I chain-related protein A, the ligand for the activating NKG2D receptor expressed on NK- and cytotoxic T-cells. Concomitantly, ΔPK also upregulated the secretion of inflammatory cytokines TNF-α, granulocyte macrophage colony-stimulating factor and IL-1β through autophagy-mediated activation of Toll-like receptor 2 pathways and pyroptosis, and it inhibited the expression of the negative immune checkpoint regulator cytotoxic T-lymphocyte antigen 4. Pharmacologic inhibition of these processes significantly reduces the oncolytic activity of ΔPK.
Collapse
Affiliation(s)
- Dominique Bollino
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Aric Colunga
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Baiquan Li
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Laure Aurelian
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
17
|
Sun Y, Jin X, Liu X, Zhang M, Liu W, Li Z, Han N, Tan M, Chi D, Zhu J, Zhang B, Yu B, Wu J. MicroRNA let-7i regulates dendritic cells maturation targeting interleukin-10 via the Janus kinase 1-signal transducer and activator of transcription 3 signal pathway subsequently induces prolonged cardiac allograft survival in rats. J Heart Lung Transplant 2015; 35:378-388. [PMID: 26755202 DOI: 10.1016/j.healun.2015.10.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/25/2015] [Accepted: 10/31/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND In this study, we investig1ated whether microRNA let-7i regulates dendric cell maturation targeting interleukin-10 (IL-10) via the Janus kinase 1-signal transducer and activator of transcription 3 (JAK1-STAT3) signal pathway subsequently prolongs rat cardiac allograft survival. METHODS Quantitative real-time reverse transcriptase polymerase chain reaction, enzyme linked immunosorbent assay, and dual-luciferase assay were performed to verify whether IL-10 was the target of let-7i, and regulatory T cells were assessed by flow cytometry and immunohistochemical study. Western blot was performed to detect JAK1, STAT3, and phosphorylated STAT3 expression. Lewis recipients of Dark Agouti hearts were transfused with phosphate-buffered saline, lipopolysaccharide (LPS)-mature dendritic cells (mDCs), or let-7i-inhibitor-mDCs. Allograft survival times were recorded, and histologic studies were performed. RESULTS Expression of IL-10 messenger RNA level and production of IL-10 were increased in let-7i-inhibitor-mDCs compared with LPS-mDCs. Luciferase activity showed that the translational level of the IL-10 luciferase reporter was decreased by let-7i mimic but increased by let-7i-inhibitor. MicroRNA let-7i inhibitor suppressed DC maturation; however, pretreatment of IL-10 small interfering RNA attenuated the suppression. Expression of JAK1, STAT3, and phosphorylated STAT3 in mDCs were suppressed by let-7i mimic, and pre-treatment of IL-10 small interfering RNA, however, were upregulated by let-7i inhibitor. Lewis recipients transfused with let-7i-inhibitor-mDCs significantly prolonged Dark Agouti cardiac allograft survival. The allografts transfused with let-7i-inhibitor-mDCs showed slight cell infiltration and significantly preserved graft structure. Inhibition of let-7i increased CD4(+)CD25(+)forkhead box P3(+) regulatory T cells and modulated cytokine profiles in vivo and in vitro. CONCLUSIONS MicroRNA let-7i regulated DC maturation and function targeting IL-10 through the JAK1-STAT3 pathway. Moreover, transfusion of LPS-induced mDCs transfected with let-7i inhibitor induced prolonged cardiac allograft survival and generated regulatory T cells.
Collapse
Affiliation(s)
- Yong Sun
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangyuan Jin
- Department of Thoracic Surgery, the Third Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xianglan Liu
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Maomao Zhang
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Liu
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenchao Li
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Na Han
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Miaoxin Tan
- Department of Cardiology, the First Hospital of Fangshan District, Beijing, China
| | - Di Chi
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingyi Zhu
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Zhang
- Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Yu
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jian Wu
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China; Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
18
|
Mackern-Oberti JP, Vega F, Llanos C, Bueno SM, Kalergis AM. Targeting dendritic cell function during systemic autoimmunity to restore tolerance. Int J Mol Sci 2014; 15:16381-417. [PMID: 25229821 PMCID: PMC4200801 DOI: 10.3390/ijms150916381] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/29/2014] [Accepted: 09/05/2014] [Indexed: 12/11/2022] Open
Abstract
Systemic autoimmune diseases can damage nearly every tissue or cell type of the body. Although a great deal of progress has been made in understanding the pathogenesis of autoimmune diseases, current therapies have not been improved, remain unspecific and are associated with significant side effects. Because dendritic cells (DCs) play a major role in promoting immune tolerance against self-antigens (self-Ags), current efforts are focusing at generating new therapies based on the transfer of tolerogenic DCs (tolDCs) during autoimmunity. However, the feasibility of this approach during systemic autoimmunity has yet to be evaluated. TolDCs may ameliorate autoimmunity mainly by restoring T cell tolerance and, thus, indirectly modulating autoantibody development. In vitro induction of tolDCs loaded with immunodominant self-Ags and subsequent cell transfer to patients would be a specific new therapy that will avoid systemic immunosuppression. Herein, we review recent approaches evaluating the potential of tolDCs for the treatment of systemic autoimmune disorders.
Collapse
Affiliation(s)
- Juan P Mackern-Oberti
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Portugal 49, Santiago 8330025, Chile.
| | - Fabián Vega
- Departamento de Inmunología Clínica y Reumatología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 350, Santiago 8330033, Chile.
| | - Carolina Llanos
- Departamento de Inmunología Clínica y Reumatología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 350, Santiago 8330033, Chile.
| | - Susan M Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Portugal 49, Santiago 8330025, Chile.
| | - Alexis M Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Portugal 49, Santiago 8330025, Chile.
| |
Collapse
|
19
|
Du J, Zhang J, Zou D, Ye L, Chen GG, Yang C. Effects of interleukin-10 gene deficiency on hepatic biochemical metabolism in mice. Clin Exp Med 2014; 15:321-5. [PMID: 24925637 DOI: 10.1007/s10238-014-0291-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 04/22/2014] [Indexed: 10/25/2022]
Abstract
The aim of this study was to investigate the effect of interleukin-10 (IL-10) gene deficiency on mouse liver function. The experimental mice were divided into wild-type and IL-10 knockout groups. Serological biomarkers for liver functions were detected by the automatic biochemical analyzer AU5400. The pathological changes were assessed by the light microscope. The levels of inducible nitric oxide synthase (iNOS) and interleukin-1β (IL-1β) in liver tissues were determined by quantitative real-time PCR and enzyme-linked immunosorbent assay. Compared with the wild type, the serum levels of albumin (ALB), total protein, total bilirubin and direct bilirubin in IL-10-deficient mice were significantly decreased (P < 0.05). No obvious pathological changes including liver necrosis and inflammatory cell infiltration were found. The expression of iNOS and IL-1β genes, the serum levels of iNOS and IL-1β were significantly higher in IL-10-deficient mice than in wild-type mice (P < 0.05). The absence of IL-10 gene can significantly decrease serum ALB and bilirubin. The effect may be related to the upregulation of iNOS and IL-1β.
Collapse
Affiliation(s)
- Jing Du
- Department of Clinical Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | | | | | | | | | | |
Collapse
|