1
|
Cui X, Liu W, Jiang H, Zhao Q, Hu Y, Tang X, Liu X, Dai H, Rui H, Liu B. IL-12 family cytokines and autoimmune diseases: A potential therapeutic target? J Transl Autoimmun 2025; 10:100263. [PMID: 39759268 PMCID: PMC11697604 DOI: 10.1016/j.jtauto.2024.100263] [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: 10/20/2024] [Revised: 11/28/2024] [Accepted: 12/01/2024] [Indexed: 01/07/2025] Open
Abstract
In recent years, the discovery of IL-12 family cytokines, which includes IL-12, IL-23, IL-27, IL-35, and IL-39, whose biological functions directly or indirectly affect various autoimmune diseases. In autoimmune diseases, IL-12 family cytokines are aberrantly expressed to varying degrees. These cytokines utilize shared subunits to influence T-cell activation and differentiation, thereby regulating the balance of T-cell subsets, which profoundly impacts the onset and progression of autoimmune diseases. In such conditions, IL-12 family members are aberrantly expressed to varying degrees. By exploring their immunomodulatory functions, researchers have identified varying therapeutic potentials for each member. This review examines the physiological functions of the major IL-12 family members and their interactions, discusses their roles in several autoimmune diseases, and summarizes the progress of clinical studies involving monoclonal antibodies targeting IL-12 and IL-23 subunits currently available for treatment.
Collapse
Affiliation(s)
- Xiaoyu Cui
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
| | - Wu Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Hanxue Jiang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Qihan Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
| | - Yuehong Hu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
| | - Xinyue Tang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
| | - Xianli Liu
- Shunyi Branch, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100310, China
| | - Haoran Dai
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Shunyi Branch, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100310, China
| | - Hongliang Rui
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Baoli Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
2
|
Bracamonte-Baran W, Kim ST. The Current and Future of Biomarkers of Immune Related Adverse Events. Immunol Allergy Clin North Am 2025; 45:223-249. [PMID: 40287170 DOI: 10.1016/j.iac.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
With their groundbreaking clinical responses, immune checkpoint inhibitors (ICIs) have ushered in a new chapter in cancer therapeutics. However, they are often associated with life-threatening or organ-threatening autoimmune/autoinflammatory phenomena, collectively termed immune-related adverse events (irAEs). In this review, we will first describe the mechanisms of action of ICIs as well as irAEs. Next, we will review biomarkers for predicting the development of irAEs or stratifying risks.
Collapse
Affiliation(s)
- William Bracamonte-Baran
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA
| | - Sang T Kim
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA.
| |
Collapse
|
3
|
Tao P, Su B, Mao X, Lin Y, Zheng L, Zou X, Yang H, Liu J, Li H. Interleukin-35 inhibits NETs to ameliorate Th17/Treg immune imbalance during the exacerbation of cigarette smoke exposed-asthma via gp130/STAT3/ferroptosis axis. Redox Biol 2025; 82:103594. [PMID: 40101533 PMCID: PMC11964675 DOI: 10.1016/j.redox.2025.103594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025] Open
Abstract
Cigarette smoke (CS) exposure amplifies neutrophil accumulation. IL-35, a novel cytokine with anti-inflammatory properties, is involved in protection against asthma. However, the biological roles of neutrophils and the precise molecular mechanisms of IL-35 in CS exposed-asthma remain unclear. We showed that the exacerbation of CS exposed-asthma leads to dramatically increased neutrophil counts and an imbalance in DC-Th17/Treg immune responses. RNA sequencing revealed that NETs, part of a key biological process in neutrophils, were significantly upregulated in the context of CS exposed-asthma exacerbation and that IL-35 treatment downregulated NET-associated gene expression. Targeted degradation of NETs, rather than neutrophil depletion, alleviated the CS exposed-asthma. Mechanistically, STAT3 phosphorylation promoted ferroptosis, exacerbating NET release, which in turn enhanced dendritic cell (DC) antigen presentation, activated T cells, and specifically promoted Th17 cell differentiation while inhibiting Treg cells. IL-35 acting on the gp130 receptor alleviated STAT3-mediated ferroptosis-associated NET formation. In summary, our study revealed a novel mechanism by which IL-35 inhibited NET formation, subsequently alleviating neutrophilic inflammation and restoring the DC-Th17/Treg imbalance in CS exposed-asthma, highlighting the potential of IL-35 as a targeted therapeutic strategy.
Collapse
Affiliation(s)
- Peizhi Tao
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Beiting Su
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xueyan Mao
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yusen Lin
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li Zheng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaoling Zou
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hailing Yang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jing Liu
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China.
| | - Hongtao Li
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Diseases of Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
4
|
Wang J, Hao Y, Yang Y, Zhang Y, Xu C, Yang R. Gut microbiota derived indole-3-acetic acid ameliorates precancerous inflammatory intestinal milieu to inhibit tumorigenesis through IL-35. J Immunother Cancer 2025; 13:e011155. [PMID: 40274281 PMCID: PMC12020765 DOI: 10.1136/jitc-2024-011155] [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: 11/25/2024] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Gut microbiota can significantly alter the risk or progression of cancer by maintaining gut immune system homeostasis. However, the exact mechanism by which the gut microbiota and its metabolites influence colorectal tumorigenesis is unclear. METHODS The roles of tryptophan metabolite indole-3-acetic acid (IAA) in inflammation and tumor development were investigated in dextran sodium sulfate (DSS) and azoxymethane (AOM)-DSS mouse models with or without IAA supplementation and with or without Lactobacillus reuteri-produced IAA. Pregnane X receptor (PXR) knockout (KO) mice and aryl hydrocarbon receptor KO mice were used to explore the mechanism by which IAA regulates interleukin (IL)-35 expression. IL-35+ immune cells were stimulated in vitro and analyzed by flow cytometry. Additionally, metabolites were analyzed by liquid chromatography-mass spectrometry. RESULTS We found that IAA, a metabolite of tryptophan produced in the gut by L. reuteri, can inhibit the development of colitis by inducing IL-35 expression in immunosuppressant cells. HuREG3αIECtg mice had high levels of intestinal microbiota-derived IAA, and these mice were resistant to AOM-DSS-induced cancer. Patients with colorectal cancer also had low peripheral blood levels of IAA. Further studies revealed that IAA-producing L. reuteri alleviated colitis symptoms and inhibited colon tumors by inducing macrophages, T cells, and B cells to produce IL-35. Finally, PXR KO completely abolished the effects of IAA on immune cells. CONCLUSION We demonstrate that gut microbiota-derived IAA can improve the precancerous colon inflammatory environment through IL-35, thereby inhibiting tumorigenesis, suggesting that IAA may be a preventive factor for colitis-related cancers.
Collapse
Affiliation(s)
| | - Yang Hao
- Nankai University School of Medicine, Tianjin, China
| | - Yazheng Yang
- Nankai University School of Medicine, Tianjin, China
| | - Yuan Zhang
- Nankai University School of Medicine, Tianjin, China
| | - Chen Xu
- Nankai University, Tianjin, China
| | - Rongcun Yang
- Nankai University Medical School, Nankai University School of Medicine, Tianjin, China
| |
Collapse
|
5
|
Kiani L, Momtahan M, Shiravani Z, Ghaderi A, Fattahi MJ, Moradi Alamdarloo S, Hashemi A, Arman R, Mosavat SH. Role of serum IL-35 levels in patients with benign and malignant primary ovarian tumors: a case-control study. Sci Rep 2025; 15:12827. [PMID: 40229348 PMCID: PMC11997060 DOI: 10.1038/s41598-025-97349-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 04/03/2025] [Indexed: 04/16/2025] Open
Abstract
IL-35 is known to enhance tumor progression by promoting angiogenesis, increasing cancer cell proliferation, and facilitating immune suppression. Elevated levels of IL-35 have been correlated with the severity of malignancy and clinical stage in various cancers. The aim of this study was to investigate the relationship between serum IL-35 levels and the presence and clinicopathological features of primary benign and malignant ovarian tumors. This case-control study consisted of 60 women diagnosed with primary benign and malignant ovarian tumors and 60 age-matched healthy controls. The demographic and clinicopathological data were also collected. The serum level of IL-35 was evaluated using an ELISA kit, and the results were analyzed using SPSS software. The patients had a mean age of 47.93 ± 14.52 which was similar to that of the controls (53.43 ± 11.69) (P = 0.519). The mean serum level of IL-35 was 7.01 ± 0.843 pg/ml in the control group and 7.24 ± 0.811 pg/ml in the patient group (P = 0.213). There was no significant difference in serum IL-35 levels between the tumor types (P = 0.991). There was also no significant association between serum IL-35 levels and disease stage (P = 0.559), grade (P = 0.635), lymph node involvement (P = 0.091), or tumor size (P = 0.564). No significant difference in serum IL-35 levels was observed between the patient and control groups, nor was there a significant association between IL-35 levels and tumor characteristics (stage, grade, size, lymph node involvement. However, further studies with more cases at different stages of the disease are necessary.
Collapse
Affiliation(s)
- Leila Kiani
- Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mozhdeh Momtahan
- Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Shiravani
- Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Maternal-fetal Medicine Research Center, Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Javad Fattahi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Shaghayegh Moradi Alamdarloo
- Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Maternal-fetal Medicine Research Center, Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Atefeh Hashemi
- Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Maternal-fetal Medicine Research Center, Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razieh Arman
- Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Hamdollah Mosavat
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
6
|
Chen Q, Zhang W, Xie T, Dong J, Zhang J, Zha L. Genetic variant rs2243115 of the IL-12/IL-35 pathway contributes to the risk of coronary artery disease. Int J Med Sci 2025; 22:2155-2164. [PMID: 40303485 PMCID: PMC12035836 DOI: 10.7150/ijms.102562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Coronary artery disease (CAD) involves inflammation. IL-12p35, a common subunit of both IL-12 and IL-35, is encoded by the IL12A gene and is a potential therapeutic target in CAD. We probed into the genetic relationships between IL12A and CAD in a Chinese Han population to provide a novel potential target and a theoretical basis for the anti-inflammatory therapies in CAD. Materials and Methods: In total, 768 patients with CADs and 768 controls were recruited for a case-control association analysis of the functional genetic variant rs2243115 of IL12A. Allelic and genotypic associations between rs2243115 and CAD and its subgroup were assessed by Logistic regression analysis. Additionally, multiple linear regression analysis was performed to explore the association between rs2243115, serum lipid levels and CAD severity. Bioinformatic tools were used to predict the potential function of rs2243115. Results: Our results showed no differences in the allele and genotype frequency distribution of rs2243115 between patients with CAD and controls. The subgroup analysis found no association between rs2243115 and CAD in either male or female groups. Furthermore, rs2243115 was not related to early- or late-onset CAD, or CAD severity. However, we did observe that rs2243115 was negatively related to HDL-c level (P=0.016, β =-0.063) and positively related to LDL-c level (P=0.029, β=0.058). Biological function prediction indicated many functional elements in the rs2243115 region, suggesting that rs2243115 may regulate gene expression in the IL-12/IL-35 pathway. Conclusion: The functional genetic variant, rs2243115, of IL12A, may play a role in CAD by regulating the IL-12/IL-35 pathway and affecting lipid levels and inflammatory responses, thereby providing a potential therapeutic target for CAD.
Collapse
Affiliation(s)
- Qianwen Chen
- Department of Pediatric Cardiology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenjuan Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Geriatrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tian Xie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiangtao Dong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Junxia Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Lingfeng Zha
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
7
|
Wang P, Li Y, Zhao L, Liu B, Cai Z, Zhang P, Li P, Gao X, Zhan Y. High interleukin-35 expression is associated with the severity of rheumatic mitral stenosis. Front Immunol 2025; 16:1537497. [PMID: 40264784 PMCID: PMC12011801 DOI: 10.3389/fimmu.2025.1537497] [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: 12/01/2024] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
Background Rheumatic mitral stenosis (RMS) is the most common manifestation of rheumatic heart disease, with high morbidity and mortality. Interleukin-35 (IL-35) is a novel anti-inflammatory cytokine associated with many autoimmune diseases. However, the relation between IL-35 expression and RMS remains unknown. We aimed to study IL-35 expression in RMS and its association with disease progression. Methods IL-35 concentration was analyzed in blood samples from 40 patients, including 20 moderate, 20 severe RMS, and 20 healthy controls by ELISA. Mitral valve (MV) IL-35 expression was determined by western blot and immunohistochemistry in patients with RMS (22 and 29 cases, respectively) in comparison to control specimens with mitral valve prolapsed (5 cases, respectively). Results IL-35 levels were significantly elevated in the blood of the RMS patients compared to those from healthy subjects(p<0.05) and positively correlated with the severity of RMS (r=0.317, p<0.05). The expression of IL-35 and its subunits (p35 and EBI3) was also detected in MV tissues of patients with moderate or severe RMS. The expression of IL-35 and its subunits (p35 and EBI3) had a positive association with the severity of RMS in MV tissues (r=0.528, p<0.01; r=0.561, p<0.001; r=0.456, p<0.01). Co-localization of p35 and EBI3 was seen in MV tissues of RMS patients in a predominantly perivascular pattern. Conclusion We show for the first time an increase of IL-35 level in the blood and MV tissues of RMS patients, which is strongly correlated with the severity of RMS. These results suggest that IL-35 plays an important regulatory role in the progression of RMS.
Collapse
Affiliation(s)
- Ping Wang
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yaxiong Li
- Department of Cardiovascular Surgery, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Li Zhao
- Key Laboratory of Cardiovascular Research of Yunnan Province, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Bin Liu
- Key Laboratory of Cardiovascular Research of Yunnan Province, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Zhibin Cai
- Key Laboratory of Cardiovascular Research of Yunnan Province, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Peng Zhang
- Key Laboratory of Cardiovascular Research of Yunnan Province, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Peng Li
- Department of Cardiovascular Surgery, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Xuezhen Gao
- Health Examination Center, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Yong Zhan
- Division of Cardiac Surgery, Cardiovascular Center, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
8
|
Abdul-Aziz AI. Molecular study and determining the levels of some interleukins in children with Entamoeba histolytica. Cytokine 2025; 188:156890. [PMID: 39965431 DOI: 10.1016/j.cyto.2025.156890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025]
Abstract
The processes by which amebiasis causes host mucosal inflammation are not entirely understood. In order to identify Entamoeba histolytica molecularly and measure the amounts of certain interleukins in children who were infected, this study was conducted. It was conducted between April and Stamper 2024. In Medical City, Baghdad, Iraq, 290 children suffering from diarrhea who were between the ages of 1 and 13 had their stool and blood samples taken. The microscopic examination showed that 128(44.1 %) children out of 290 were infected with Entamoeba spp. while when the samples were examined using the RT-PCR technique, it was found that the number of positive samples was 53(36.6 %) for E. histolytica The percentage of infected male children was 41.5 % while, the percentage of female children was 58.5 %. For interleukins, IL-2 was significantly increased in infected children (265.02 ± 11.78) compared with control (89.36 ± 5.19 pg/ml). IL-4 was significantly increased in infected children (249.11 ± 12.47 pg/ml) compared with control (54.84 ± 1.93 pg/ml). IL-6 was significantly increased in infected children (7.28 ± 1.57 pg/ml) compared with control (2.11 ± 0.42 pg/ml). IL-17 was significantly increased (P < 0.05) in infected children (15.46 ± 1.93 pg/ml) compared with control (7.06 ± 0.67 pg/ml). According to this research, PCR technology is a more sensitive and accurate way to diagnose E. histolytica than the direct method with an optical microscope. Inconclusion, this study found that children with E. histolytica had considerably increased serum levels of several interleukins, including IL-2, IL-4, IL-6, and IL-17.
Collapse
Affiliation(s)
- Ayser I Abdul-Aziz
- Department of Medical laboratory techniques/Collage of Health and medical technologies, Al-Turath University, Iraq.
| |
Collapse
|
9
|
Jie F, Dong F, Xu L, Deng S, Wang Q, Wu Q. Cytokine Expression and Cytolytic Effect of Natural Killer Cells are Suppressed in Septic Shock. Scand J Immunol 2025; 101:e70023. [PMID: 40254928 DOI: 10.1111/sji.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 02/21/2025] [Accepted: 04/04/2025] [Indexed: 04/22/2025]
Abstract
Septic shock is the most severe stage of sepsis. How immune dysregulation contributes to the pathogenesis of septic shock has not been thoroughly understood. In the current research, the phenotype and function of circulating natural killer (NK) cells of septic patients were characterised. The absolute number of NK cells was comparably reduced in septic shock survivors and non-survivors, probably owing to elevated NK cell apoptosis. Activating receptors including signalling lymphocytic activation molecule 4 (SLAMF4), natural killer cell p30-related protein (NKp30), natural killer group 2, member D (NKG2D), and DNAX accessory molecule 1 (DNAM-1) were significantly downregulated on NK cell surface in septic shock patients, especially non-survivors. Furthermore, the patients' NK cells exhibited lower expression of granzyme B and perforin, weaker target cell-induced degranulation and cytokine expression, as well as incompetent cytolytic effect. These alterations were more profound in septic shock non-survivors. Importantly, serum interleukin-35 (IL-35), which is an immunosuppressive cytokine, was remarkably elevated in septic shock patients. Besides, serum interleukin-35 concentration was positively correlated with disease scores but negatively correlated with NK cell activating receptor expression. In vitro assays indicated IL-35-induced strong suppression of NK cell activity, as evidenced by concomitant downregulation of cytokines and activating receptors along with inhibition of cytolytic capacity. Therefore, we uncovered for the first time the contributing role of IL-35 in septic shock-related human NK cell dysfunction.
Collapse
Affiliation(s)
- Fengying Jie
- The Department of Intensive Care Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Fang Dong
- The Department of Intensive Care Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Lingwen Xu
- The Department of Intensive Care Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Shuping Deng
- The Department of Intensive Care Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Qian Wang
- The Department of Intensive Care Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Qun Wu
- The Department of Intensive Care Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| |
Collapse
|
10
|
Wardell CM, Boardman DA, Levings MK. Harnessing the biology of regulatory T cells to treat disease. Nat Rev Drug Discov 2025; 24:93-111. [PMID: 39681737 DOI: 10.1038/s41573-024-01089-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 12/18/2024]
Abstract
Regulatory T (Treg) cells are a suppressive subset of CD4+ T cells that maintain immune homeostasis and restrain inflammation. Three decades after their discovery, the promise of strategies to harness Treg cells for therapy has never been stronger. Multiple clinical trials seeking to enhance endogenous Treg cells or deliver them as a cell-based therapy have been performed and hint at signs of success, as well as to important limitations and unanswered questions. Strategies to deplete Treg cells in cancer are also in active clinical testing. Furthermore, multi-dimensional methods to interrogate the biology of Treg cells are leading to a refined understanding of Treg cell biology and new approaches to harness tissue-specific functions for therapy. A new generation of Treg cell clinical trials is now being fuelled by advances in nanomedicine and synthetic biology, seeking more precise ways to tailor Treg cell function. This Review will discuss recent advances in our understanding of human Treg cell biology, with a focus on mechanisms of action and strategies to assess outcomes of Treg cell-targeted therapies. It highlights results from recent clinical trials aiming to enhance or inhibit Treg cell activity in a variety of diseases, including allergy, transplantation, autoimmunity and cancer, and discusses ongoing strategies to refine these approaches.
Collapse
Affiliation(s)
- Christine M Wardell
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dominic A Boardman
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Megan K Levings
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
11
|
Zidlik V, Hurnik P, Vantuchova Y, Michalcova S, Skarda J, Hulinova T, Purova D, Ehrmann J. FOXP3, IL-35, and PD-L1 in intra- and peritumoral lymphocytic infiltrate of cutaneous melanomas as an important part of antitumor immunity. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2025. [PMID: 39865911 DOI: 10.5507/bp.2024.040] [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: 01/28/2025] Open
Abstract
BACKGROUND The tumor microenvironment is a significant mediator enabling tumor growth and progression. Tumor-infiltrating lymphocytes (TILs) are an important component of this but tumor cells develop mechanisms by which they can escape the action of the immune system. Immunosuppressive mechanisms cooperate with each other and involve cells of the immune system, the tumor microenvironment itself, chemokines and cytokines. In this study, we examined the FOXP3+, IL-35+, and PD-L1+ lymphocytes in tumor tissues as they are contributing to immunosuppression in some tumors, including melanoma. Such cells are also associated with tumor progression, early metastasis, and prognosis. METHODS AND RESULTS In this study, 95 cutaneous melanomas and 25 melanocytic nevi as a control group were examined by immunohistochemistry for FOXP3+, IL-35+, and PD-L1+ lymphocytes. Melanomas were divided into four groups according to the TNM classification: pT1 (35), pT2 (21), pT3 (21), and pT4 (18). PD-L1+ lymphocytes were enriched in pT3- and pT4-stage melanomas, especially in the periphery of the lesions (P<0.001). The number of FOXP3+ lymphocytes was positively correlated with the stage of the disease, especially in the center of the tumors (P<0.001). Likewise, IL-35+ lymphocytes (P<0.001) were enriched with the stage of the tumor. CONCLUSION This article demonstrates that the immunosuppressive environment develops in proportion to the stage of the melanoma. The most significant changes are found at the tumor periphery, confirming the heterogeneity of the tumor stroma which is more pronounced in more advanced tumors and which may contribute to the greater aggressiveness in these peripheral zones.
Collapse
Affiliation(s)
- Vladimir Zidlik
- Institute of Pathology and Molecular Genetics, Faculty Hospital Ostrava, Ostrava, Czech Republic
- Institute of Clinical and Molecular Pathology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Pavel Hurnik
- Institute of Pathology and Molecular Genetics, Faculty Hospital Ostrava, Ostrava, Czech Republic
- Institute of Clinical and Molecular Pathology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Yvetta Vantuchova
- Department of Dermatology, Faculty Hospital Ostrava, Ostrava, Czech Republic
| | - Simona Michalcova
- Department of Dermatology, Faculty Hospital Ostrava, Ostrava, Czech Republic
| | - Jozef Skarda
- Institute of Pathology and Molecular Genetics, Faculty Hospital Ostrava, Ostrava, Czech Republic
- Institute of Clinical and Molecular Pathology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Tereza Hulinova
- Institute of Pathology and Molecular Genetics, Faculty Hospital Ostrava, Ostrava, Czech Republic
- Institute of Clinical and Molecular Pathology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Dana Purova
- Social Health Institute, Palacky University Olomouc, Olomouc, Czech Republic
| | - Jiri Ehrmann
- Institute of Pathology and Molecular Genetics, Faculty Hospital Ostrava, Ostrava, Czech Republic
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| |
Collapse
|
12
|
Zhang Y, Cheng F, Cai X, Wu J. Malignant behaviors and immune response in melanoma: Epstein-Barr virus induced gene 3 as a therapeutic target based on an in-vitro exploration. PeerJ 2024; 12:e18730. [PMID: 39726752 PMCID: PMC11670768 DOI: 10.7717/peerj.18730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
Background Epstein-Barr virus induced gene 3 (EBI3), a member of the IL-12 family, is known to be involved in malignant progression in a variety of cancers, but its role in melanoma is unclear. The aim of this study was to explore the effects of EBI3 on the malignant phenotype melanoma to reveal its potential as a therapeutic target. Methods In this study, we used bioinformatics to analyze the expression of EBI3 in pan-cancer and verified its expression level in melanoma cells by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Subsequently, the effects of EBI3 knockdown on cell proliferation, migration and invasion were detected using the Cell Counting Kit-8 (CCK-8) and Transwell assays. Changes in immune-related cytokines were detected by ELISA, and macrophage polarization was observed using immunofluorescence. Finally, the phosphorylation levels of signaling pathways such as Smad3, STAT6 and cGAS-STING were analyzed by Western blot. Results EBI3 was evidently highly-expressed in melanoma, and silencing of EBI3 could visibly suppress the survival and migration/invasion of melanoma cells, concurrent with the increased levels of BAX and CDH1 and the decreased expressions of BCL2 and CDH2. Meanwhile, EBI3 knockdown diminished the phosphorylation levels of both Smad3 and STAT6 and the levels of immune response-relevant cytokines in melanoma cells, while aggravating the macrophage M1 polarization and the expression of cGAS, p-STING and p-IRE1 α in THP-1 monocyte-derived macrophages co-cultured with EBI3-silenced melanoma cells. Conclusion This study filled the blank on the involvement of EBI3 in melanoma, hinting the possibility of controlling EBI3 as a therapeutic strategy in the management of melanoma.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Medical Aesthetics, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fengrui Cheng
- Department of Medical Aesthetics, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xingrui Cai
- Surgery of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingping Wu
- Department of Medical Aesthetics, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
13
|
Ji L, Mei X, Yuan W, Guo H, Zhang Y, Zhang Z, Zou Y, Liu Y, Zhu H, Qian Z, Shen Y. Plasma Interleukin-35 Levels Predict the Prognosis in Patients with HBV-Related Acute-on-Chronic Liver Failure. Viruses 2024; 16:1960. [PMID: 39772266 PMCID: PMC11680333 DOI: 10.3390/v16121960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
This study aimed to investigate the impact of IL-35 on the prognosis of patients with HBV-ACLF. We recruited 69 patients with HBV-ACLF, 20 patients with chronic hepatitis B (CHB), 17 patients with liver cirrhosis (LC), and 20 healthy controls (HCs) from a regional infectious disease treatment center in China. Plasma levels of IL-35 at baseline were detected using ELISA. Plasma IL-35 levels in the HBV-ACLF group were the highest among all four groups. Furthermore, survivors exhibited significantly higher IL-35 levels than non-survivors (p < 0.001). IL-35 levels correlated with MELD (r = -0.678, p < 0.001), COSSH-ACLF IIs (r = -0.581, p < 0.001), alpha-fetoprotein (AFP) (r = 0.433, p < 0.001), creatinine (Cr) (r =-0.396, p = 0.001), and lactate (r =-0.38, p =0.001). The combination of plasma IL-35 and MELD score had the highest mortality prediction efficiency, with an area under the curve (AUC) of 0.895 (95% CI: 0.812-0.978, p < 0.001), a sensitivity of 80.6%, and a specificity of 93.9%. Additionally, the Kaplan-Meier analysis revealed that lower levels of IL-35 (≤191.5pg/mL) were associated with poorer survival rates in HBV-ACLF patients (p < 0.001). Our results demonstrated that IL-35 could be an effective predictive marker for the prognosis of HBV-ACLF and improve the predictive performance when combined with the MELD score.
Collapse
Affiliation(s)
- Liujuan Ji
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (L.J.); (X.M.); (W.Y.); (H.G.); (Y.Z.); (Z.Z.); (Y.Z.); (Y.L.); (H.Z.)
| | - Xue Mei
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (L.J.); (X.M.); (W.Y.); (H.G.); (Y.Z.); (Z.Z.); (Y.Z.); (Y.L.); (H.Z.)
| | - Wei Yuan
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (L.J.); (X.M.); (W.Y.); (H.G.); (Y.Z.); (Z.Z.); (Y.Z.); (Y.L.); (H.Z.)
| | - Hongying Guo
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (L.J.); (X.M.); (W.Y.); (H.G.); (Y.Z.); (Z.Z.); (Y.Z.); (Y.L.); (H.Z.)
| | - Yuyi Zhang
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (L.J.); (X.M.); (W.Y.); (H.G.); (Y.Z.); (Z.Z.); (Y.Z.); (Y.L.); (H.Z.)
| | - Zhengguo Zhang
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (L.J.); (X.M.); (W.Y.); (H.G.); (Y.Z.); (Z.Z.); (Y.Z.); (Y.L.); (H.Z.)
| | - Ying Zou
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (L.J.); (X.M.); (W.Y.); (H.G.); (Y.Z.); (Z.Z.); (Y.Z.); (Y.L.); (H.Z.)
| | - Yu Liu
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (L.J.); (X.M.); (W.Y.); (H.G.); (Y.Z.); (Z.Z.); (Y.Z.); (Y.L.); (H.Z.)
| | - Hui Zhu
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (L.J.); (X.M.); (W.Y.); (H.G.); (Y.Z.); (Z.Z.); (Y.Z.); (Y.L.); (H.Z.)
| | - Zhiping Qian
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (L.J.); (X.M.); (W.Y.); (H.G.); (Y.Z.); (Z.Z.); (Y.Z.); (Y.L.); (H.Z.)
| | - Yinzhong Shen
- Department of Infection and Immunity, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| |
Collapse
|
14
|
Stajer M, Horacek JM, Kupsa T, Zak P. The role of chemokines and interleukins in acute lymphoblastic leukemia: a systematic review. J Appl Biomed 2024; 22:165-184. [PMID: 40033805 DOI: 10.32725/jab.2024.024] [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: 08/10/2024] [Accepted: 11/15/2024] [Indexed: 03/05/2025] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood hematological malignancy, but it also affects adult patients with worse prognosis and outcomes. Leukemic cells benefit from protective mechanisms, which are mediated by intercellular signaling molecules - cytokines. Through these signals, cytokines modulate the biology of leukemic cells and their surroundings, enhancing the proliferation, survival, and chemoresistance of the disease. This ultimately leads to disease progression, refractoriness, and relapse, decreasing the chances of curability and overall survival of the patients. Targeting and modulating these pathological processes without affecting the healthy physiology is desirable, offering more possibilities for the treatment of ALL patients, which still remains unsatisfactory in certain cases. In this review, we comprehensively analyze the existing literature and ongoing trials regarding the role of chemokines and interleukins in the biology of ALL. Focusing on the functional pathways, genetic background, and critical checkpoints, we constructed a summary of molecules that are promising for prognostic stratification and mainly therapeutic use. Targeted therapy, including chemokine and interleukin pathways, is a new and promising approach to the treatment of cancer. With the expansion of our knowledge, we are able to uncover a spectrum of new potential checkpoints in order to modulate the disease biology. Several cytokine-related targets are advancing toward clinical application, offering the hope of higher disease response rates to treatment.
Collapse
Affiliation(s)
- Martin Stajer
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Jan M Horacek
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Tomas Kupsa
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Pavel Zak
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| |
Collapse
|
15
|
Chandwaskar R, Dalal R, Gupta S, Sharma A, Parashar D, Kashyap VK, Sohal JS, Tripathi SK. Dysregulation of T cell response in the pathogenesis of inflammatory bowel disease. Scand J Immunol 2024; 100:e13412. [PMID: 39394898 DOI: 10.1111/sji.13412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 10/14/2024]
Abstract
Inflammatory bowel disease (IBD), comprised of Crohn's disease (CD) and ulcerative colitis (UC), are gut inflammatory diseases that were earlier prevalent in the Western Hemisphere but now are on the rise in the East, with India standing second highest in the incidence rate in the world. Inflammation in IBD is a cause of dysregulated immune response, wherein helper T (Th) cell subsets and their cytokines play a major role in the pathogenesis of IBD. In addition, gut microbiota, environmental factors such as dietary factors and host genetics influence the outcome and severity of IBD. Dysregulation between effector and regulatory T cells drives gut inflammation, as effector T cells like Th1, Th17 and Th9 subsets Th cell lineages were found to be increased in IBD patients. In this review, we attempted to discuss the role of different Th cell subsets together with other T cells like CD8+ T cells, NKT and γδT cells in the outcome of gut inflammation in IBD. We also highlighted the potential therapeutic candidates for IBD.
Collapse
Affiliation(s)
- Rucha Chandwaskar
- Amity Institute of Microbial Technology (AIMT), Amity University Jaipur, Rajasthan, India
| | - Rajdeep Dalal
- Infection and Immunology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, India
| | - Saurabh Gupta
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Aishwarya Sharma
- Sri Siddhartha Medical College and Research Center, Tumkur, Karnataka, India
| | - Deepak Parashar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vivek K Kashyap
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
| | - Jagdip Singh Sohal
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Subhash K Tripathi
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, Washington, USA
| |
Collapse
|
16
|
Naji NS, Sathish M, Karantanos T. Inflammation and Related Signaling Pathways in Acute Myeloid Leukemia. Cancers (Basel) 2024; 16:3974. [PMID: 39682161 DOI: 10.3390/cancers16233974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic malignancy, and inflammatory signaling is involved in its pathogenesis. Cytokines exert a robust effect on the progression of AML and affect survival outcomes. The dysregulation in the cytokine network may foster a pro-tumorigenic microenvironment, increasing leukemic cell proliferation, decreasing survival and driving drug resistance. The dominance of pro-inflammatory mediators such as IL-11β, TNF-α and IL-6 over anti-inflammatory mediators such as TGF-β and IL-10 has been implicated in tumor progression. Additionally, inflammatory cytokines have favored certain populations of hematopoietic stem and progenitor cells with mutated clonal hematopoiesis genes. This article summarizes current knowledge about inflammatory cytokines and signaling pathways in AML, their modes of action and the implications for immune tolerance and clonal hematopoiesis, with the aim of finding potential therapeutic interventions to improve clinical outcomes in AML patients.
Collapse
Affiliation(s)
- Nour Sabiha Naji
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Mrudula Sathish
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Theodoros Karantanos
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
17
|
Wang X, Tang J, Zhang X, Zeng H. The Protective Effect of Esculentoside A on MPL/lpr Mice by Upregulating the Expression of CD19+IL-35+Breg Cells and Interleukin-35. Int Arch Allergy Immunol 2024; 186:358-368. [PMID: 39500292 DOI: 10.1159/000541812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 09/27/2024] [Indexed: 03/27/2025] Open
Abstract
INTRODUCTION Esculentoside A (EsA) is one of the main components of the traditional Chinese medicine Phytolacca esculenta. The possible mechanism of action of EsA in the treatment of lupus nephritis (LN) was explored by observing the effects of EsA on CD19+ IL-35+regulatory B (IL-35+Breg) cells. METHODS Twenty-four MRL/lpr mice were randomly divided into control, EsA, and EsA+IL-12p35 antibody groups. Mice were administered the respective treatments intraperitoneally once a day for 4 weeks. The urine protein/creatinine ratio (UPCR) and blood creatinine (Cr) and IL-35, IL-10, and IL-17 expression levels were measured. Body and spleen weight were measured to calculate the splenic index (SI). Flow cytometry was performed to determine the proportion of CD19+ IL-35+ Breg cells in the spleen. Hematoxylin-eosin and PASM-Masson staining of renal tissues were performed, and the "Austin" acute index (AI) system for LN was determined. RESULTS The most severe conditions were seen in mice in the control group, with the highest UPCR, Cr, and IL-17 levels and SI and AI scores; the most severe renal histopathology, and the lowest proportion of CD19+ IL-35+ Breg cells and IL-35 and IL-10 levels. This was followed by the EsA+IL-12p35 antibody group. The EsA group had the lowest UPCR, Cr, and IL-17 levels and SI and AI scores; the mildest renal lesions; and the highest proportion CD19+ IL-35+ Breg cells and IL-35 and IL-10 levels. CONCLUSION EsA delayed the progression of LN by promoting the proliferation of CD19+ IL-35+ Breg cells, upregulating the expression of IL-35, and decreasing the secretion of IL-17.
Collapse
Affiliation(s)
- Xing Wang
- Department of Nephrology and Rheumatology, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China,
| | - Jieyin Tang
- Department of Nephrology and Rheumatology, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China
| | - Xianggui Zhang
- Department of Nephrology and Rheumatology, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China
| | - Huilin Zeng
- Department of Nephrology and Rheumatology, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, China
| |
Collapse
|
18
|
van der Mescht MA, Steel HC, de Beer Z, Masenge A, Abdullah F, Ueckermann V, Anderson R, Rossouw TM. T-Cell Phenotypes and Systemic Cytokine Profiles of People Living with HIV Admitted to Hospital with COVID-19. Microorganisms 2024; 12:2149. [PMID: 39597537 PMCID: PMC11596914 DOI: 10.3390/microorganisms12112149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/29/2024] Open
Abstract
Whether SARS-CoV-2 infection leads to a higher mortality and morbidity in people living with HIV (PLWH) in Africa remains inconclusive. In this study, we explored the differences in the T-cell phenotypes between people with and without HIV on the day of admission (V1) and ±7 days later (V2), as well as their cytokine/chemokine profiles on V1. Patients admitted with COVID-19 were recruited between May 2020 and December 2021 from the Steve Biko Academic and Tshwane District Hospitals in Pretoria, South Africa. Of 174 patients, 37 (21%) were PLWH. T-cell profiles were determined by flow cytometry, and cytokine levels were determined using a multiplex suspension bead array. PLWH were significantly younger than those without HIV, and were more likely to be female. In an adjusted analysis, PLWH had higher percentages of CD4+ central memory (CM) programmed cell death protein 1 (PD-1)+, CD8+ effector memory (EM)2, and CD8+ EM4 CD57+ cells, as well as higher concentrations of interleukin (IL)-35 at admission. PLWH with CD4+ T-cell counts of >200 cells/mm3 had altered CD4+ and CD8+ T-cell profiles, lower levels of systemic inflammation measured by plasma ferritin and PCT levels, and less severe disease. PLWH with CD4+ T-cell counts of <200 cells/mm3 on admission had higher concentrations of IL-6 and lower levels of IL-29. At V2, the percentages of CD4+ CM PD-1+ T-cells and CD8+ EM4 T-cells co-expressing CD57 and PD-1 remained higher in PLWH, while all other CD8+ EM populations were lower. Fewer CD8+ EM T-cells after ±7 days of admission may be indicative of mechanisms inhibiting EM T-cell survival, as indicated by the higher expression of IL-35 and the T-cell maturation arrest observed in PLWH. This profile was not observed in PLWH with severe immunodeficiency, highlighting the need for differentiated care in the broader PLWH population.
Collapse
Affiliation(s)
- Mieke A. van der Mescht
- Department of Immunology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (M.A.v.d.M.); (H.C.S.); (Z.d.B.); (R.A.)
| | - Helen C. Steel
- Department of Immunology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (M.A.v.d.M.); (H.C.S.); (Z.d.B.); (R.A.)
| | - Zelda de Beer
- Department of Immunology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (M.A.v.d.M.); (H.C.S.); (Z.d.B.); (R.A.)
- Tshwane District Hospital, Pretoria 0084, South Africa
| | - Andries Masenge
- Department of Statistics, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria 0001, South Africa;
| | - Fareed Abdullah
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0001, South Africa; (F.A.); (V.U.)
- Office of AIDS and TB Research, South African Medical Research Council, Pretoria 0001, South Africa
- Department of Public Health Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Veronica Ueckermann
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0001, South Africa; (F.A.); (V.U.)
| | - Ronald Anderson
- Department of Immunology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (M.A.v.d.M.); (H.C.S.); (Z.d.B.); (R.A.)
| | - Theresa M. Rossouw
- Department of Immunology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (M.A.v.d.M.); (H.C.S.); (Z.d.B.); (R.A.)
| |
Collapse
|
19
|
Huang Q, Zhu J. Regulatory T cell-based therapy in type 1 diabetes: Latest breakthroughs and evidence. Int Immunopharmacol 2024; 140:112724. [PMID: 39098233 DOI: 10.1016/j.intimp.2024.112724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/10/2024] [Accepted: 07/16/2024] [Indexed: 08/06/2024]
Abstract
Autoimmune diseases (ADs) are among the most significant health complications, with their incidence rising in recent years. Type 1 diabetes (T1D), an AD, targets the insulin-producing β cells in the pancreas, leading to chronic insulin deficiency in genetically susceptible individuals. Regulatory immune cells, particularly T-cells (Tregs), have been shown to play a crucial role in the pathogenesis of diabetes by modulating immune responses. In diabetic patients, Tregs often exhibit diminished effectiveness due to various factors, such as instability in forkhead box P3 (Foxp3) expression or abnormal production of the proinflammatory cytokine interferon-gamma (IFN-γ) by autoreactive T-cells. Consequently, Tregs represent a potential therapeutic target for diabetes treatment. Building on the successful clinical outcomes of chimeric antigen receptor (CAR) T-cell therapy in cancer treatment, particularly in leukemias, the concept of designing and utilizing CAR Tregs for ADs has emerged. This review summarizes the findings on Treg targeting in T1D and discusses the benefits and limitations of this treatment approach for patients suffering from T1D.
Collapse
Affiliation(s)
- Qiongxiao Huang
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China
| | - Jing Zhu
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
20
|
Chakraborty R, Mukherjee AK, Bala A. Interleukin-35: A key player managing pre-diabetes and chronic inflammatory type 1 autoimmune diabetes. World J Diabetes 2024; 15:2147-2151. [PMID: 39493554 PMCID: PMC11525726 DOI: 10.4239/wjd.v15.i10.2147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/16/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Interleukin-35 (IL-35) is a novel protein comprising IL-12α and IL-27β chains. The IL12A and EBI3 genes are responsible for its production. The study of IL-35 has experienced a substantial increase in interest in recent years, as demonstrated by many research papers. Recent clinical studies have shown that individuals who do not have a C-peptide have notably reduced amounts of IL-35 in their blood serum. This is accompanied by a drop in the percentage of IL-35+ Treg cells, regulatory B cells, and CD8+ FOXP3+ cells that produce IL-35. This article em-phasizes the potential significance of IL-35 expression in governing the immune response and its involvement in chronic inflammatory autoimmune diabetes in pancreatic inflammation. It demonstrates IL-35's ability to regulate cytokine proportions, modulate B cells, and protect against autoimmune diabetes. However, further investigation is necessary to ascertain the precise mechanism of IL-35, and meticulous planning is essential for clinical studies.
Collapse
Affiliation(s)
- Ratul Chakraborty
- Life Sciences Division, Institute of Advanced Study in Science and Technology, Guwahati 781035, Guwahati, Assam, India
| | - Ashis Kumar Mukherjee
- Life Sciences Division, Institute of Advanced Study in Science and Technology, Guwahati 781035, Guwahati, Assam, India
| | - Asis Bala
- Pharmacology and Drug Discovery Research Laboratory, Division of Life Sciences, Institute of Advanced Study in Science and Technology, Guwahati 781035, Assam, India
| |
Collapse
|
21
|
Hadifar S, Masoudzadeh N, Heydari H, Mashayekhi Goyonlo V, Kerachian M, Daneshpazhooh M, Sadeghnia A, Tootoonchi N, Erfanian Salim R, Rafati S, Harandi AM. Intralesional gene expression profile of JAK-STAT signaling pathway and associated cytokines in Leishmania tropica-infected patients. Front Immunol 2024; 15:1436029. [PMID: 39364404 PMCID: PMC11446769 DOI: 10.3389/fimmu.2024.1436029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024] Open
Abstract
Background The JAK-STAT signaling pathway is a central cascade of signal transduction for the myriad of cytokines in which dysregulation has been implicated in progression of inflammatory and infectious diseases. However, the involvement of this pathway in human cutaneous leishmaniasis (CL) due to Leishmania (L.) tropica warrants further investigation. Methods This study sought to investigate differential gene expression of several cytokines and their associated jak-stat genes in the lesions of L. tropica-infected patients byquantitative Real-Time PCR. Further, the expression of five inhibitory immune checkpoint genes was evaluated. Results Results showed that the gene expression levelsof both Th1 (ifng, il12, il23) and Th2 (il4, il10) types cytokines were increased in the lesion of studied patients. Further, elevated expression levels of il35, il21, il27 and il24 genes were detected in the lesions of CL patients. Notably, the expression of the majority of genes involved in JAK/STAT signaling pathway as well as checkpoint genes including pdl1, ctla4 and their corresponding receptors was increased. Conclusion Our finding revealed dysregulation of cytokines and related jak-stat genes in the lesion of CL patients. These results highlight the need for further exploration of the functional importance of these genes in the pathogenesis of, and immunity to, CL.
Collapse
Affiliation(s)
- Shima Hadifar
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Nasrin Masoudzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Hossein Heydari
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | | | - Mohammadali Kerachian
- Cutaneous Leishmaniasis Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Daneshpazhooh
- Autoimmune Bullous Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Ali M. Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
22
|
Chakraborty R, Mukherjee AK, Bala A. Breakthroughs in road mapping IL-35 mediated immunotherapy for type-1 and autoimmune diabetes mellitus. Cytokine 2024; 181:156692. [PMID: 38986251 DOI: 10.1016/j.cyto.2024.156692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/22/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024]
Abstract
IL-35 is a recently discovered protein made up of IL-12α and IL-27β chains. It is encoded by IL12A and EBI3 genes. Interest in researching IL-35 has significantly increased in recent years, as evidenced by numerous scientific publications. Diabetes is on the rise globally, causing more illness and death in developing countries. The International Diabetes Federation (IDF) reports that diabetes is increasingly affecting children and teenagers, with varying rates across different regions. Therefore, scientists seek new diabetes treatments despite the growth of drug research. Recent research aims to emphasize IL-35 as a critical regulator of diabetes, especially type 1 and autoimmune diabetes. This review provides an overview of recent research on IL-35 and its link to diabetes and its associated complications. Studies suggest that IL-35 can offer protection against type-1 diabetes and autoimmune diabetes by regulating macrophage polarization, T-cell-related cytokines, and regulatory B cells (Bregs). This review will hopefully assist biomedical scientists in exploring the potential role of IL-35-mediated immunotherapy in treating diabetes. However, further research is necessary to determine the exact mechanism and plan clinical trials.
Collapse
Affiliation(s)
- Ratul Chakraborty
- Pharmacology and Drug Discovery Research Laboratory, Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Guwahati 781035, Assam, India; Academy of Scientific and Innovative Research (AcSIR), AcSIR (an Indian Institute of National Importance), Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201002, India
| | - Ashis K Mukherjee
- Microbial Biotechnology and Protein Research Laboratory, Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Guwahati 781035, Assam, India
| | - Asis Bala
- Pharmacology and Drug Discovery Research Laboratory, Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Guwahati 781035, Assam, India; Academy of Scientific and Innovative Research (AcSIR), AcSIR (an Indian Institute of National Importance), Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
23
|
Chandra DJ, Alber B, Saultz JN. The Immune Resistance Signature of Acute Myeloid Leukemia and Current Immunotherapy Strategies. Cancers (Basel) 2024; 16:2615. [PMID: 39123343 PMCID: PMC11311077 DOI: 10.3390/cancers16152615] [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: 05/03/2024] [Revised: 06/29/2024] [Accepted: 07/11/2024] [Indexed: 08/12/2024] Open
Abstract
Acute myeloid leukemia (AML) is a complex hematopoietic clonal disorder with limited curative options beyond stem cell transplantation. The success of transplant is intimately linked with the graft versus leukemia effect from the alloreactive donor immune cells including, T and NK cells. The immune system plays a dynamic role in leukemia survival and resistance. Despite our growing understanding of the immune microenvironment, responses to immune-based therapies differ greatly between patients. Herein, we review the biology of immune evasion mechanisms in AML, discuss the current landscape of immunotherapeutic strategies, and discuss the implications of therapeutic targets. This review focuses on T and NK cell-based therapy, including modified and non-modified NK cells, CAR-T and CAR-NK cells, antibodies, and checkpoint blockades. Understanding the complex interchange between immune tolerance and the emergence of tumor resistance will improve patient outcomes.
Collapse
Affiliation(s)
- Daniel J. Chandra
- Division of Hematology/Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA;
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA;
| | - Bernhard Alber
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA;
| | - Jennifer N. Saultz
- Division of Hematology/Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA;
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA;
| |
Collapse
|
24
|
Elmasry MF, Obaid YA, El-Samanoudy SI, Nour ZA, Doss SS. Estimation of the tissue and serum levels of IL-35 in Mycosis fungoides: a case-control study. Arch Dermatol Res 2024; 316:349. [PMID: 38850434 PMCID: PMC11162372 DOI: 10.1007/s00403-024-03115-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 04/06/2024] [Accepted: 04/26/2024] [Indexed: 06/10/2024]
Abstract
Mycosis fungoides (MF) is the most common primary cutaneous T-cell lymphoma (CTCL) with its etiology not yet fully understood. Interleukin (IL)-35 is an inhibitory cytokine that belongs to the IL-12 family. Elevated IL-35 in the plasma and the tumor microenvironment increases tumorigenesis and indicates poor prognosis in different types of malignancies. The objective of this study is to estimate the expression levels of IL-35 in tissue and serum of MF patients versus healthy controls. This case-control study included 35 patients with patch, plaque, and tumor MF as well as 30 healthy controls. Patients were fully assessed, and serum samples and lesional skin biopsies were taken prior to starting treatment. The IL-35 levels were measured in both serum and tissue biopsies by ELISA technique. Both tissue and serum IL-35 levels were significantly higher in MF patients than in controls (P < 0.001) and tissue IL-35 was significantly higher than serum IL-35 in MF patients (P < 0.001). Tissue IL-35 was significantly higher in female patients and patients with recurrent MF compared to male patients and those without recurrent disease (P < 0.001). Since both tissue and serum IL-35 levels are increased in MF, IL-35 is suggested to have a possible role in MF pathogenesis. IL-35 can be a useful diagnostic marker for MF. Tissue IL-35 can also be an indicator of disease recurrence.
Collapse
Affiliation(s)
- Maha Fathy Elmasry
- Dermatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | | - Zeinab Ahmed Nour
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Sally Sameh Doss
- Dermatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
25
|
Wei X, Zhang J, Cui J, Xu W, Zhao G, Guo C, Yuan W, Zhou X, Ma J. Adaptive plasticity of natural interleukin-35-induced regulatory T cells (Tr35) that are required for T-cell immune regulation. Theranostics 2024; 14:2897-2914. [PMID: 38773985 PMCID: PMC11103508 DOI: 10.7150/thno.90608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 04/09/2024] [Indexed: 05/24/2024] Open
Abstract
Background: IL-35 potently inhibits immune responses both in vivo and in vitro. However, the specific characteristics of IL-35-producing cells, including their developmental origin, cellular phenotype, and function, are unknown. Methods: By using a novel IL-35 reporter mouse (Ebi3-Dre-Thy1.1) and double transgenic fate-mapping reporter mice (35EbiT-Rosa26-rox-tdTomato reporter mice or Foxp3 fate-mapping system), we tracked and analyzed the differentiation and developmental trajectories of Tr35 cells in vivo. And then we investigated the therapeutic effects of OVA-specific Tr35 cells in an OVA-induced allergic airway disease model. Results: We identified a subset of cells, denoted Tr35 cells, that secrete IL-35 but do not express Foxp3. These cells have high expression of molecules associated with T-cell activation and can inhibit T-cell proliferation in vitro. Our analyses showed that Tr35 cells are a distinct subpopulation of cells that are independent of Tr1 cells. Tr35 cells exhibit a unique gene expression profile and tissue distribution. The presence of Thy1.1 (Ebi3) expression in Tr35 cells indicates their active secretion of IL-35. However, the proportion of ex-Tr35 cells (Thy1.1-) is significantly higher compared to Tr35 cells (Thy1.1+). This suggests that Tr35 cells possess the ability to regulate IL-35 expression rapidly in vivo. Tr35 cells downregulated the expression of the inflammatory cytokines IL-4, IFN-γ and IL-17A. However, once Tr35 cells lost IL-35 expression and became exTr35 cells, the expression of inflammatory cytokines was upregulated. Importantly, our findings indicate that Tr35 cells have therapeutic potential. In an OVA-induced allergic airway disease mouse model, Tr35 cell reinfusion significantly reduced airway hyperresponsiveness and histopathological airway and lung inflammation. Conclusions: We have identified a subset of Tregs, Tr35 cells, that are distinct from Tr1 cells. Tr35 cells can dynamically regulate the secretion of inflammatory cytokines by controlling IL-35 expression to regulate inflammatory immune responses.
Collapse
Affiliation(s)
- Xundong Wei
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Guangxi Nanning, P.R. China
| | - Jianhua Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing 100101, P.R. China
| | - Jian Cui
- Department of General Surgery, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, P.R. China
| | - Wei Xu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing 100101, P.R. China
| | - Gang Zhao
- Department of General Surgery, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, P.R. China
| | - Chang Guo
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Wei Yuan
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P.R. China
| | - Xuyu Zhou
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing 100101, P.R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, P.R. China
| | - Jie Ma
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Guangxi Nanning, P.R. China
| |
Collapse
|
26
|
Bracamonte-Baran W, Kim ST. The Current and Future of Biomarkers of Immune Related Adverse Events. Rheum Dis Clin North Am 2024; 50:201-227. [PMID: 38670721 PMCID: PMC11232920 DOI: 10.1016/j.rdc.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
With their groundbreaking clinical responses, immune checkpoint inhibitors (ICIs) have ushered in a new chapter in cancer therapeutics. However, they are often associated with life-threatening or organ-threatening autoimmune/autoinflammatory phenomena, collectively termed immune-related adverse events (irAEs). In this review, we will first describe the mechanisms of action of ICIs as well as irAEs. Next, we will review biomarkers for predicting the development of irAEs or stratifying risks.
Collapse
Affiliation(s)
- William Bracamonte-Baran
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA
| | - Sang T Kim
- Department of Rheumatology, Allergy & Immunology, Yale University, 300 Cedar Street, TAC S541, New Haven, CT 06520, USA.
| |
Collapse
|
27
|
Khalaf AK, Sabeeh FA. Immunological and cytokine profile (IL-25 and IL-35) in patients with Entamoeba histolytica infection in Southern Iraq. ARCHIVES OF RAZI INSTITUTE 2024; 79:315-320. [PMID: 39463702 PMCID: PMC11512188 DOI: 10.32592/ari.2024.79.2.315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/06/2023] [Indexed: 10/29/2024]
Abstract
Amoebiasis is an intestinal disease caused by a unicellular parasite called Entamoeba histolytica. Interleukin-35 (IL-35) is the youngest specific member of the IL-12 family that plays a major role in the inhibitory function of regulatory T cells (Tregs) to curb inflammatory responses. IL-25 of the IL-17 family, which is widely released by Th2 cells and epithelial cells, is a warning signal produced upon cell or tissue injury to activate immune cells. The present study aimed to determine the cytokine profile (IL-25 and IL-35) in patients with E. histolytica infection in southern Iraq. This hospital-based study was conducted from August 2022 to May 2023. The study participants were patients with E. histolytica infection admitted to the infection department of general hospitals in Thi-qar province, southern Iraq. Initially, E. histolytica amebiasis was detected in the patients by nested multiplex PCR. All collected sera were tested with the Human Interleukin 35 (Biotech, China, Cat.RD-IL35-Hu) and IL25 (Biotech, China, Cat.RD-IL25-Hu) ELISA kits according to the instructions of the manufacturer. A total of 80 patients, including 50 patients with E. histolytica infection and 30 subjects in the control group without E. histolytica infection, were enrolled in the present study. The results showed a significant difference (p<0.001) in the serum level of IL-25 in patients with E. histolytica infection (4275.19 pg/mL) compared to individuals in the control group without E. histolytica infection (2186 pg/mL). Statistical analysis showed that there was no significant difference in the serum levels of IL-35 patients with E. histolytica infection compared with individuals in the control group without E. histolytica infection. The results of the present study show that the level of IL-25 is high in patients with E. histolytica infection. This indicates the important role of IL-25 in the activation of the immune system during intestinal inflammation. Therefore, this cytokine can be used as a diagnostic marker for E. histolytica infection.
Collapse
Affiliation(s)
- AKh Khalaf
- Department of Microbiology, College of Medicine, University of Thi-qar, Thi-qar, Iraq
| | - FAb Sabeeh
- Department of Microbiology, College of Medicine, University of Thi-qar, Thi-qar, Iraq
| |
Collapse
|
28
|
Cao Y, Liu YL, Lu XY, Kai HL, Han Y, Zheng YL. Integrative analysis from multi-center studies identifies a weighted gene co-expression network analysis-based Tregs signature in ovarian cancer. ENVIRONMENTAL TOXICOLOGY 2024; 39:736-750. [PMID: 37713585 DOI: 10.1002/tox.23948] [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: 06/17/2023] [Revised: 07/31/2023] [Accepted: 08/13/2023] [Indexed: 09/17/2023]
Abstract
Ovarian cancer (OC) is a malignancy associated with poor prognosis and has been linked to regulatory T cells (Tregs) in the immune microenvironment. Nevertheless, the association between Tregs-related genes (TRGs) and OC prognosis remains incompletely understood. The xCell algorithm was used to analyze Tregs scores across multiple cohorts. Weighted gene co-expression network analysis (WGCNA) was utilized to identify potential TRGs and molecular subtypes. Furthermore, we used nine machine learning algorithms to create risk models with prognostic indicators for patients. Reverse transcription-quantitative polymerase chain reaction and immunofluorescence staining were used to demonstrate the immunosuppressive ability of Tregs and the expression of key TRGs in clinical samples. Our study found that higher Tregs scores were significantly correlated with poorer overall survival. Recurrent patients exhibited increased Tregs infiltration and reduced CD8+ T cell. Moreover, molecular subtyping using seven key TRGs revealed that subtype B exhibited higher enrichment of multiple oncogenic pathways and had a worse prognosis. Notably, subtype B exhibited high Tregs levels, suggesting immune suppression. In addition, we validated machine learning-derived prognostic models across multiple platform cohorts to better distinguish patient survival and predict immunotherapy efficacy. Finally, the differential expression of key TRGs was validated using clinical samples. Our study provides novel insights into the role of Tregs in the immune microenvironment of OC. We identified potential therapeutic targets derived from Tregs (CD24, FHL2, GPM6A, HOXD8, NAP1L5, REN, and TOX3) for personalized treatment and created a machining learning-based prognostic model for OC patients, which could be useful in clinical practice.
Collapse
Affiliation(s)
- Yang Cao
- Department of Obstetrics and Gynecology, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, China
| | - Ying-Lei Liu
- Department of Obstetrics and Gynecology, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, China
| | - Xiao-Yan Lu
- Department of Obstetrics and Gynecology, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, China
| | - Hai-Li Kai
- Department of Obstetrics and Gynecology, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, China
| | - Yun Han
- Department of Obstetrics and Gynecology, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, China
| | - Yan-Li Zheng
- Department of Obstetrics and Gynecology, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, China
| |
Collapse
|
29
|
Yi P, Yu W, Xiong Y, Dong Y, Huang Q, Lin Y, Du Y, Hua F. IL-35: New Target for Immunotherapy Targeting the Tumor Microenvironment. Mol Cancer Ther 2024; 23:148-158. [PMID: 37988561 DOI: 10.1158/1535-7163.mct-23-0242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/15/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023]
Abstract
Interleukin 35(IL-35) is a newly discovered inhibitory cytokine of the IL12 family. More recently, IL-35 was found to be increased in the tumor microenvironment (TME) and peripheral blood of many patients with cancer, indicating that it plays an important role in the TME. Tumors secrete cytokines that recruit myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Treg) into the TME to promote malignant progression, which is a great challenge for cancer treatment. Radiotherapy causes serious adverse effects, and tumor resistance to immune checkpoint inhibitors is still an unsolved challenge. Thus, new cancer therapy approaches are urgently needed. Numerous studies have shown that IL-35 can recruit immunosuppressive cells to enable tumor immune escape by promoting the conversion of immune cells into a tumor growth-promoting phenotype as well as facilitating tumor angiogenesis. IL-35-neutralizing antibodies were found to boost the chemotherapeutic effect of gemcitabine and considerably reduce the microvascular density of pancreatic cancer in mice. Therefore, targeting IL-35 in the TME provides a promising cancer treatment target. In addition, IL-35 may be used as an independent prognostic factor for some tumors in the near future. This review intends to reveal the interplay of IL-35 with immune cells in the TME, which may provide new options for the treatment of cancer.
Collapse
Affiliation(s)
- Pengcheng Yi
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Wenjun Yu
- Fuzhou First People's Hospital of Jiangxi Province, Fuzhou City, Jiangxi Province, P.R. China
| | - Yanhong Xiong
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yao Dong
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Qiang Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yunfei Du
- Department of Anesthesiology, Nanchang Central Hospital, Nanchang, Jiangxi, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| |
Collapse
|
30
|
Baydoun H, Camirand G. IL-35 Therapy Boosts Treg in Transplantation. Transplantation 2024; 108:14-15. [PMID: 37464475 PMCID: PMC10796832 DOI: 10.1097/tp.0000000000004708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Affiliation(s)
- Hussein Baydoun
- The Thomas E. Starzl Transplantation Institute, Departments of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh PA
| | - Geoffrey Camirand
- The Thomas E. Starzl Transplantation Institute, Departments of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh PA
| |
Collapse
|
31
|
Su X, Yang Y, Gao Y, Wang J, Hao Y, Zhang Y, Yang R. Gut microbiota CLA and IL-35 induction in macrophages through Gαq/11-mediated STAT1/4 pathway: an animal-based study. Gut Microbes 2024; 16:2437253. [PMID: 39636005 PMCID: PMC11622586 DOI: 10.1080/19490976.2024.2437253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/05/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
Gut microbiota/metabolites not only participate in the food and energy metabolism but also contribute to the host immune response and homeostasis. The alternation of gut microbiota/metabolites has been widely related to intestinal and extra-intestinal disorders such as intestinal bowel diseases (IBDs). Bactericidal substances from gut epithelial cells can regulate the composition of gut microbiota. However, the effects of regenerating protein 4 (REG4) (human)/(Reg4) (mice), a potentially bactericidal substance from gut epithelial cells, on the gut immune homeostasis maintain elusive. Here, we found that REG4/Reg4 is essential in maintaining gut immune homeostasis through REG4/Reg4 associated gut microbiota. Reg4 knockout (KO) mice were highly sensitive to DSS-mediated colitis, whereas human REG4 intestine epithelial cell transgenic (huREG4IECtg) mice exhibited more resistance to DSS-mediated colitis. Mechanistically, sequencing of gut microbiota and liquid chromatography-mass spectrometry showed that REG4/Reg4 could affect the composition of gut microbiota. REG4/Reg4 associated gut microbiota such as Lactobacillus could metabolize linoleic acid (LA) into conjugated linoleic acid (CLA). Immunoprecipitation and immunoblot showed that CLA could effectively promote the expression of IL-35 in macrophages through Gαq/11 mediated activation STAT1/4. Thus, our results demonstrate that REG4/Reg4 plays a critical role in maintaining gut immune homeostasis through CLA-mediated IL-35+ macrophages.
Collapse
Affiliation(s)
- Xiaomin Su
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yazheng Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yunhuan Gao
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Juanjuan Wang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yang Hao
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yuan Zhang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- Translational Medicine Institute, Tianjin Union Medical Center of Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| |
Collapse
|
32
|
Huang A, Liu K, Yin Z, Liu J, Wei H, Xing S, Qu Y, Huang L, Li L, Li C, Zhang L, Li X, Zheng C, Liu Q, Jiang K. IL-35 Stabilizes Treg Phenotype to Protect Cardiac Allografts in Mice. Transplantation 2024; 108:161-174. [PMID: 37464473 PMCID: PMC10718222 DOI: 10.1097/tp.0000000000004707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND Interleukin-35 (IL-35), secreted by regulatory T cells (Treg) and B cells, is immunosuppressive under both physiological and pathological conditions. However, the role of IL-35 in all responses has yet to be investigated. Here, we demonstrate that IL-35 protects allografts by stabilizing the Treg phenotype and suppressing CD8 + T-cell activation in a mouse heart transplantation model. METHODS The effect of IL-35 on immune cell infiltration in grafts and secondary lymphoid organs was examined using mass cytometry, flow cytometry, and immunofluorescence. Moreover, using quantitative real-time polymerase chain reaction, flow cytometry, and phospho-flow assays, we demonstrated that IL-35 maintains Treg phenotypes to restrain CD8 + T cells via the gp130/signal transducer and activator of transcription 1 pathway. RESULTS Mass cytometry analysis of intragraft immune cells showed that IL-35 decreased CD8 + T-cell infiltration and increased Foxp3 and IL-35 expressions in Treg. In vitro, we demonstrated that IL-35 directly promoted Treg phenotypic and functional stability and its IL-35 secretion, generating a positive feedback loop. However, Treg are required for IL-35 to exert its suppressive effect on CD8 + T cells in vitro. After depleting Treg in the recipient, IL-35 did not prolong graft survival or decrease CD8 + T-cell infiltration. Mechanistically, we found that IL-35 sustained Treg stability via the gp130/signal transducer and activator of transcription 1 signaling pathway. CONCLUSIONS Our findings highlight that IL-35 stabilizes the Treg phenotype to ameliorate CD8 + T-cell infiltration in the allograft, which has never been described in the transplanted immunological milieu.
Collapse
Affiliation(s)
- Ai Huang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kewei Liu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyi Yin
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Jie Liu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Hongyan Wei
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Shijie Xing
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Qu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Huang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liancheng Li
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chang Li
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Lei Zhang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Xiaoshi Li
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Cunni Zheng
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Quan Liu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China
| | - Ke Jiang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
33
|
Zysk W, Sitko K, Tukaj S, Zaryczańska A, Trzeciak M. Altered Gene Expression of IL-35 and IL-36α in the Skin of Patients with Atopic Dermatitis. Int J Mol Sci 2023; 25:404. [PMID: 38203575 PMCID: PMC10779293 DOI: 10.3390/ijms25010404] [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: 11/17/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
A comprehensive understanding of atopic dermatitis (AD) pathogenesis is desired, especially in the current era of novel biologics and small molecule drugs. In recent years, new cytokines have emerged that may play a significant role in the pathogenesis of AD. Using the tape stripping (TS) method, this study analyzed the gene expression of IL-35 and IL-36α in lesional and nonlesional AD skin compared with healthy skin and their association with the clinical features of AD among the Polish population. Ten AD patients and seven healthy individuals were enrolled. The lesional skin of the AD patients showed significantly higher expression levels of IL-35 compared to healthy skin (p = 0.0001). The expression level of IL-36α was significantly higher in lesional AD skin than in nonlesional AD skin (p = 0.0039) and healthy skin (p = 0.0045). There was a significant negative correlation between AD severity and the expression level of IL-35 in both lesional (R = -0.66, p = 0.048) and nonlesional skin (R = -0.9, p = 0.0016). In summary, both IL-35 and IL-36α appear to play a role in the pathogenesis of AD. Furthermore, it might be speculated that IL-35 and IL-36α may be potential candidates for disease biomarkers. However, further studies are needed to verify these assumptions and comprehensively elucidate their importance in the pathogenesis of AD.
Collapse
Affiliation(s)
- Weronika Zysk
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, 80-214 Gdansk, Poland; (W.Z.); (A.Z.)
| | - Krzysztof Sitko
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland; (K.S.); (S.T.)
| | - Stefan Tukaj
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland; (K.S.); (S.T.)
| | - Anna Zaryczańska
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, 80-214 Gdansk, Poland; (W.Z.); (A.Z.)
| | - Magdalena Trzeciak
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, 80-214 Gdansk, Poland; (W.Z.); (A.Z.)
| |
Collapse
|
34
|
Lin F, Yu H, Zhang L, Zhou J, Cao Y, Wu S, Wang J. Differential expression of interleukin-35 receptor distinguishes different subsets of granulocyte-macrophage-colony-stimulating factor-producing T helper cells in a mouse endometriosis model. Mol Immunol 2023; 164:28-38. [PMID: 37944204 DOI: 10.1016/j.molimm.2023.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/04/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023]
Abstract
The immune system contributes to the pathophysiology of endometriosis. The role of ThGM cells, which produce granulocyte macrophage-colony-stimulating factor (GM-CSF), in the pathogenesis of endometriosis remains unknown. To analyze the features of ThGM cells in endometriosis, a mouse endometriosis model was established. ThGM cells in the spleen, peritoneal fluid (PF), and endometriotic lesions (EL) were measured by flow cytometry, based on the expression of surface markers and intracellular proteins. Live ThGM cells were sorted according to chemokine receptor expression profiles and their effects on other CD4+ T cell subsets were determined by co-culture assays. An adoptive transfer assay was performed to characterize the effect of ThGM cells on endometriosis. We found that ThGM cells were present in endometriotic PF and EL. Live EL ThGM cells were enriched in CD4+CXCR3-CCR8-CCR4+CCR10+ T cells. EL ThGM cells differentially express interleukin-35 receptor (IL-35R), consisting of an IL-35R+ subset and an IL-35R- subset. The IL-35R+ subset expressed less GM-CSF, interleukin-2 (IL-2), and tumor necrosis factor-alpha (TNF-α) and proliferated slower than the IL-35R- subset. Meanwhile, the IL-35R+ subset was weaker than the IL-35R- subset in promoting the functions of Th1 and Th17 cells. ThGM cell transfer did not influence EL development but significantly alleviated pro-inflammatory cytokines in PF and ELs. Interleukin-35 (IL-35), the ligand of IL-35R, suppressed ThGM cell function and proliferation in an IL-35R-dependent manner. In summary, ThGM cells in the PF and ELs might exacerbate endometriotic inflammation. IL-35 might suppress the function of ThGM cells via IL-35R.
Collapse
Affiliation(s)
- Fengqin Lin
- The Department of Obstetrics and Gynecology at Affiliated Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province 443000, China
| | - Hongbo Yu
- The Department of Obstetrics and Gynecology at Affiliated Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province 443000, China
| | - Li Zhang
- The Department of Obstetrics and Gynecology at Affiliated Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province 443000, China
| | - Jing Zhou
- The Department of Obstetrics and Gynecology at Affiliated Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province 443000, China
| | - Yuan Cao
- The Department of Obstetrics and Gynecology at Affiliated Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province 443000, China
| | - Songli Wu
- The Department of Obstetrics and Gynecology at Affiliated Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province 443000, China
| | - Junjie Wang
- The Department of Obstetrics and Gynecology at Affiliated Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province 443000, China.
| |
Collapse
|
35
|
Li S, Su L, Luan Q, Liu G, Zeng W, Yu X. Regulatory B cells induced by interleukin-35 inhibit inflammation and alveolar bone resorption in ligature-induced periodontitis. J Periodontol 2023; 94:1376-1388. [PMID: 37086023 DOI: 10.1002/jper.23-0038] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/31/2023] [Accepted: 04/13/2023] [Indexed: 04/23/2023]
Abstract
BACKGROUND Regulatory B cells (Bregs) have been reported to suppress immune responses and alveolar bone loss in murine periodontitis models. These cells could be induced by interleukin (IL)-35 which is increased upon periodontal inflammation. Thus, this study aimed to explore the role of Bregs induced by IL-35 in periodontitis. METHODS Experimental periodontitis was induced in mice by ligature. Two weeks after ligation, the test group was systemically treated with IL-35 for 1 week. Four weeks after ligation, all mice were euthanized, and alveolar bone loss was evaluated by microcomputed tomography. Cytokines associated with periodontitis were analyzed using reverse transcription-quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. Bregs in spleens, cervical lymph nodes, and periodontal tissues were detected by flow cytometry and immunofluorescence staining. RESULTS In the mouse model of periodontitis, IL-35 induced the expansion of CD1dhi CD5+ B10 cells with increased interleukin-10 (IL-10) and IL-35 production. IL-35 administration also attenuated alveolar bone loss and reduced the levels of proinflammatory cytokines in situ. CONCLUSIONS Following ligature-induced periodontitis in mice, IL-35 inhibited periodontal inflammation and alveolar bone resorption at least partially through the induction of B10 cells and IL-35+ Bregs.
Collapse
Affiliation(s)
- Shiyi Li
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Li Su
- Center of Medical and Health Analysis, Peking University, Beijing, China
| | - Qingxian Luan
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Guojing Liu
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Wenmin Zeng
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiaoqian Yu
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
36
|
Hildenbrand K, Bohnacker S, Menon PR, Kerle A, Prodjinotho UF, Hartung F, Strasser PC, Catici DA, Rührnößl F, Haslbeck M, Schumann K, Müller SI, da Costa CP, Esser-von Bieren J, Feige MJ. Human interleukin-12α and EBI3 are cytokines with anti-inflammatory functions. SCIENCE ADVANCES 2023; 9:eadg6874. [PMID: 37878703 PMCID: PMC10599630 DOI: 10.1126/sciadv.adg6874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 09/22/2023] [Indexed: 10/27/2023]
Abstract
Interleukins are secreted proteins that regulate immune responses. Among these, the interleukin 12 (IL-12) family holds a central position in inflammatory and infectious diseases. Each family member consists of an α and a β subunit that together form a composite cytokine. Within the IL-12 family, IL-35 remains particularly ill-characterized on a molecular level despite its key role in autoimmune diseases and cancer. Here we show that both IL-35 subunits, IL-12α and EBI3, mutually promote their secretion from cells but are not necessarily secreted as a heterodimer. Our data demonstrate that IL-12α and EBI3 are stable proteins in isolation that act as anti-inflammatory molecules. Both reduce secretion of proinflammatory cytokines and induce the development of regulatory T cells. Together, our study reveals IL-12α and EBI3, the subunits of IL-35, to be functionally active anti-inflammatory immune molecules on their own. This extends our understanding of the human cytokine repertoire as a basis for immunotherapeutic approaches.
Collapse
Affiliation(s)
- Karen Hildenbrand
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Sina Bohnacker
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Zentrum München, 80802 Munich, Germany
| | - Priyanka Rajeev Menon
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Anna Kerle
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Ulrich F. Prodjinotho
- Institute for Microbiology, Immunology and Hygiene, Technical University of Munich, 81675 Munich, Germany
- Center for Global Health, Technical University of Munich, 81675 Munich, Germany
| | - Franziska Hartung
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Zentrum München, 80802 Munich, Germany
| | - Patrick C. Strasser
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Dragana A. M. Catici
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Florian Rührnößl
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Martin Haslbeck
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Kathrin Schumann
- Institute for Microbiology, Immunology and Hygiene, Technical University of Munich, 81675 Munich, Germany
| | - Stephanie I. Müller
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Clarissa Prazeres da Costa
- Institute for Microbiology, Immunology and Hygiene, Technical University of Munich, 81675 Munich, Germany
- Center for Global Health, Technical University of Munich, 81675 Munich, Germany
- German Center for Infection and Research (DZIF), partner site Munich, Germany
| | - Julia Esser-von Bieren
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Zentrum München, 80802 Munich, Germany
- Department of Immunobiology, Université de Lausanne, 1066 Epalinges, Switzerland
| | - Matthias J. Feige
- Center for Functional Protein Assemblies (CPA), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| |
Collapse
|
37
|
Neubauer-Geryk J, Wielicka M, Myśliwiec M, Zorena K, Bieniaszewski L. The Relationship between TNF-a, IL-35, VEGF and Cutaneous Microvascular Dysfunction in Young Patients with Uncomplicated Type 1 Diabetes. Biomedicines 2023; 11:2857. [PMID: 37893230 PMCID: PMC10604652 DOI: 10.3390/biomedicines11102857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/15/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
The aim of this study was to analyze the relationship between immunological markers and the dysfunction of cutaneous microcirculation in young patients with type 1 diabetes. The study group consisted of 46 young patients with type 1 diabetes and no associated complications. Microvascular function was assessed with the use of nail fold capillaroscopy before and after implementing post-occlusive reactive hyperemia. This evaluation was then repeated after 12 months. Patients were divided into two subgroups according to their baseline median coverage (defined as the ratio of capillary surface area to surface area of the image area), which was established during the initial exam (coverageBASE). Additionally, the levels of several serum biomarkers, including VEGF, TNF-a and IL-35, were assessed at the time of the initial examination. HbA1c levels obtained at baseline and after a 12-month interval were also obtained. Mean HbA1c levels obtained during the first two years of the course of the disease were also analyzed. Patients with coverageBASE below 16.85% were found to have higher levels of VEGF and TNF-α, as well as higher levels of HbA1c during the first two years following diabetes diagnosis. Our results support the hypothesis that the development of diabetic complications is strongly influenced by metabolic memory and an imbalance of pro- and anti-inflammatory cytokines, regardless of achieving adequate glycemic control.
Collapse
Affiliation(s)
- Jolanta Neubauer-Geryk
- Clinical Physiology Unit, Medical Simulation Centre, Medical University of Gdańsk, 80-210 Gdansk, Poland; (M.W.); (L.B.)
| | - Melanie Wielicka
- Clinical Physiology Unit, Medical Simulation Centre, Medical University of Gdańsk, 80-210 Gdansk, Poland; (M.W.); (L.B.)
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Division of Neonatology, Ann Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Małgorzata Myśliwiec
- Department of Pediatrics, Diabetology and Endocrinology, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Katarzyna Zorena
- Department of Immunobiology and Environment Microbiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland;
| | - Leszek Bieniaszewski
- Clinical Physiology Unit, Medical Simulation Centre, Medical University of Gdańsk, 80-210 Gdansk, Poland; (M.W.); (L.B.)
| |
Collapse
|
38
|
Schifferli A, Rüfer A, Rovo A, Nimmerjahn F, Cantoni N, Holbro A, Favre G, Dirks J, Wieland A, Faeth H, Pereira R, Kühne T. Immunomodulation with romiplostim as a second-line strategy in primary immune thrombocytopenia: The iROM study. Br J Haematol 2023; 203:119-130. [PMID: 37735543 DOI: 10.1111/bjh.19074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 06/19/2023] [Indexed: 09/23/2023]
Abstract
Thrombopoietin receptor agonists (TPO-RAs) stimulate platelet production, which might restore immunological tolerance in primary immune thrombocytopenia (ITP). The iROM study investigated romiplostim's immunomodulatory effects. Thirteen patients (median age, 31 years) who previously received first-line treatment received romiplostim for 22 weeks, followed by monitoring until week 52. In addition to immunological data, secondary end-points included the sustained remission off-treatment (SROT) rate at 1 year, romiplostim dose, platelet count and bleedings. Scheduled discontinuation of romiplostim and SROT were achieved in six patients with newly diagnosed ITP, whereas the remaining seven patients relapsed. Romiplostim dose titration was lower and platelet count response was stronger in patients with SROT than in relapsed patients. In all patients, regulatory T lymphocyte (Treg) counts increased until study completion and the counts were higher in patients with SROT. Interleukin (IL)-4, IL-9 and IL-17F levels decreased significantly in all patients. FOXP3 (Treg), GATA3 (Th2) mRNA expression and transforming growth factor-β levels increased in patients with SROT. Treatment with romiplostim modulates the immune system and possibly influences ITP prognosis. A rapid increase in platelet counts is likely important for inducing immune tolerance. Better outcomes might be achieved at an early stage of autoimmunity, but clinical studies are needed for confirmation.
Collapse
Affiliation(s)
- Alexandra Schifferli
- Department of Hematology/Oncology, University Children's Hospital Basel, Basel, Switzerland
| | - Axel Rüfer
- Department of Hematology, Cantonal Hospital Lucerne, Lucerne, Switzerland
| | - Alicia Rovo
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Falk Nimmerjahn
- Department of Biology, Institute of Genetics, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Nathan Cantoni
- Department of Hematology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Andreas Holbro
- Department of Hematology, University Hospital Basel, Basel, Switzerland
| | - Geneviève Favre
- Department of Hematology, Cantonal Hospital Liestal, Switzerland
| | - Jan Dirks
- Department of Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Anna Wieland
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Heike Faeth
- Medical University of Basel, Basel, Switzerland
| | | | - Thomas Kühne
- Department of Hematology/Oncology, University Children's Hospital Basel, Basel, Switzerland
| |
Collapse
|
39
|
Reynolds L, Luo Z, Singh K. Diabetic complications and prospective immunotherapy. Front Immunol 2023; 14:1219598. [PMID: 37483613 PMCID: PMC10360133 DOI: 10.3389/fimmu.2023.1219598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
The incidence of Diabetes Mellitus is increasing globally. Individuals who have been burdened with diabetes for many years often develop complications as a result of hyperglycemia. More and more research is being conducted highlighting inflammation as an important factor in disease progression. In all kinds of diabetes, hyperglycemia leads to activation of alternative glucose metabolic pathways, resulting in problematic by-products including reactive oxygen species and advanced glycation end products. This review takes a look into the pathogenesis of three specific diabetic complications; retinopathy, nephropathy and neuropathy as well as their current treatment options. By considering recent research papers investigating the effects of immunotherapy on relevant conditions in animal models, multiple strategies are suggested for future treatment and prevention of diabetic complications with an emphasis on molecular targets associated with the inflammation.
Collapse
|
40
|
Burlingham WJ, Jankowska-Gan E, Fechner JH, Little CJ, Wang J, Hong S, Molla M, Sullivan JA, Foley DP. Extracellular Vesicle-associated GARP/TGFβ:LAP Mediates "Infectious" Allo-tolerance. Transplant Direct 2023; 9:e1475. [PMID: 37250483 PMCID: PMC10212611 DOI: 10.1097/txd.0000000000001475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 05/31/2023] Open
Abstract
Here we test the hypothesis that, like CD81-associated "latent" IL35, the transforming growth factor (TGF)β:latency-associated peptide (LAP)/glycoprotein A repetitions predominant (GARP) complex was also tethered to small extracellular vesicles (sEVs), aka exosomes, produced by lymphocytes from allo-tolerized mice. Once these sEVs are taken up by conventional T cells, we also test whether TGFβ could be activated suppressing the local immune response. Methods C57BL/6 mice were tolerized by i.p. injection of CBA/J splenocytes followed by anti-CD40L/CD154 antibody treatment on days 0, 2, and 4. On day 35, spleen and lymph nodes were extracted and isolated lymphocytes were restimulated with sonicates of CBA splenocytes overnight. sEVs were extracted from culture supernatants by ultracentrifugation (100 000g) and assayed for (a) the presence of TGFβ:LAP associated with tetraspanins CD81,CD63, and CD9 by enzyme-linked immunosorbent assay; (b) GARP, critical to membrane association of TGFβ:LAP and to activation from its latent form, as well as various TGFβ receptors; and (c) TGFβ-dependent function in 1° and 2° immunosuppression of tetanus toxoid-immunized B6 splenocytes using trans-vivo delayed-type hypersensitivity assay. Results After tolerization, CBA-restimulated lymphocytes secreted GARP/TGFβ:LAP-coated extracellular vesicles. Like IL35 subunits, but unlike IL10, which was absent from ultracentrifuge pellets, GARP/TGFβ:LAP was mainly associated with CD81+ exosomes. sEV-bound GARP/TGFβ:LAP became active in both 1° and 2° immunosuppression, the latter requiring sEV uptake by "bystander" T cells and reexpression on the cell surface. Conclusions Like other immune-suppressive components of the Treg exosome, which are produced in a latent form, exosomal GARP/TGFβ:LAP produced by allo-specific regulatory T cells undergoes either immediate activation (1° suppression) or internalization by naive T cells, followed by surface reexpression and subsequent activation (2°), to become suppressive. Our results imply a membrane-associated form of TGFβ:LAP that, like exosomal IL35, can target "bystander" lymphocytes. This new finding implicates exosomal TGFβ:LAP along with Treg-derived GARP as part of the infectious tolerance network.
Collapse
Affiliation(s)
- William J. Burlingham
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Ewa Jankowska-Gan
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - John H. Fechner
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Christopher J. Little
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Jianxin Wang
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI
| | - Seungpyo Hong
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI
| | - Miraf Molla
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Jeremy A. Sullivan
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - David P. Foley
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
41
|
Zeng W, Wang Y, Cao Y, Xing F, Yang X. Study of dietary‑induced progression of psoriasis‑like mice based on gut macrophage polarization. Exp Ther Med 2023; 25:278. [PMID: 37206572 PMCID: PMC10189756 DOI: 10.3892/etm.2023.11976] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 03/02/2023] [Indexed: 05/21/2023] Open
Abstract
The aim of the present study was to investigate the influence of stimulating food (SF), a Traditional Chinese Medicine term for a high protein, high fat diet, on psoriasis exacerbation. It was hypothesized that SF disposed psoriasis-like aggravation might be related to inflammatory pathways induction via gut dysbiosis. In the present study, mice were fed either an SF or normal diet for 4 weeks. In the last week, their back hair was removed to establish psoriasis-like dermatitis by imiquimod. After sacrifice, blood samples, alimentary tissues and skin lesions were collected and tested by enzyme-linked immunosorbent assay, western blotting, immunohistochemistry and immunofluorescence. Compared with normal diet groups, body weight and blood glucose of SF diet mice were not increased, but they exhibited higher modified Psoriasis Area and Severity Index scores and corresponding epithelial hyperproliferation. Unexpectedly, skin lesions showed abnormal lower protein expressions of Notch and TLR-2/NF-κB p65 signaling pathway, which was attributable to severe skin damage. No difference was observed in the structure and inflammatory cell infiltration of the gut between groups. Instead, macrophage polarization (M1/M2) in the gut of the SF diet group marked by high expression of CD11b (a marker of macrophage, M1) and mild low expression of MRC1 (a marker of macrophage, M2), which resulted in increased TNF-α, decreased IL-10, IL-35, and unchanged IL-17 in serum. Furthermore, serum derived from SF diet mice promoted translocation of NF-κB p65 in HaCaT cells, which indirectly suggested a systemic inflammation. These results suggested that mice fed a continuous SF diet for a time could change gut macrophage polarization, which secretes proinflammatory cytokines into blood circulation. Once transported to skin lesions, these cytokines activate psoriasis tissue resident immune cells and present as psoriasis exacerbation.
Collapse
Affiliation(s)
- Wucheng Zeng
- Department of Dermatology and Venereology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310007, P.R. China
| | - Yihan Wang
- Department of Dermatology, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Yi Cao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Hangzhou, Zhejiang 310006, P.R. China
| | - Fengling Xing
- Department of Dermatology and Venereology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310007, P.R. China
- Correspondence to: Dr Fengling Xing, Department of Dermatology and Venereology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 453 Tiyuchang Road, Hangzhou, Zhejiang 310007, P.R. China
| | - Xiaohong Yang
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Hangzhou, Zhejiang 310006, P.R. China
- Correspondence to: Dr Fengling Xing, Department of Dermatology and Venereology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 453 Tiyuchang Road, Hangzhou, Zhejiang 310007, P.R. China
| |
Collapse
|
42
|
Mahfooz NS, Merling MR, Claeys TA, Dowling JW, Forero A, Robinson RT. Human IL-35 Inhibits the Bioactivity of IL-12 and Its Interaction with IL-12Rβ2. Immunohorizons 2023; 7:431-441. [PMID: 37289499 PMCID: PMC10580122 DOI: 10.4049/immunohorizons.2300039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 06/09/2023] Open
Abstract
IL-35 is an immunosuppressive cytokine with roles in cancer, autoimmunity, and infectious disease. In the conventional model of IL-35 biology, the p35 and Ebi3 domains of this cytokine interact with IL-12Rβ2 and gp130, respectively, on the cell surface of regulatory T and regulatory B cells, triggering their suppression of Th cell activity. Here we use a human IL-12 bioactivity reporter cell line, protein binding assays, and primary human Th cells to demonstrate an additional mechanism by which IL-35 suppresses Th cell activity, wherein IL-35 directly inhibits the association of IL-12 with its surface receptor IL-12Rβ2 and downstream IL-12-dependent activities. IL-12 binding to the surface receptor IL-12Rβ1 was unaffected by IL-35. These data demonstrate that in addition to acting via regulatory T and regulatory B cells, human IL-35 can also directly suppress IL-12 bioactivity and its interaction with IL-12Rβ2.
Collapse
Affiliation(s)
- Najmus S. Mahfooz
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Marlena R. Merling
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Tiffany A. Claeys
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Jack W. Dowling
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Adriana Forero
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Richard T. Robinson
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| |
Collapse
|
43
|
Sonoda J, Mizoguchi I, Inoue S, Watanabe A, Sekine A, Yamagishi M, Miyakawa S, Yamaguchi N, Horio E, Katahira Y, Hasegawa H, Hasegawa T, Yamashita K, Yoshimoto T. A Promising Needle-Free Pyro-Drive Jet Injector for Augmentation of Immunity by Intradermal Injection as a Physical Adjuvant. Int J Mol Sci 2023; 24:ijms24109094. [PMID: 37240448 DOI: 10.3390/ijms24109094] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Current worldwide mRNA vaccination against SARS-CoV-2 by intramuscular injection using a needled syringe has greatly protected numerous people from COVID-19. An intramuscular injection is generally well tolerated, safer and easier to perform on a large scale, whereas the skin has the benefit of the presence of numerous immune cells, such as professional antigen-presenting dendritic cells. Therefore, intradermal injection is considered superior to intramuscular injection for the induction of protective immunity, but more proficiency is required for the injection. To improve these issues, several different types of more versatile jet injectors have been developed to deliver DNAs, proteins or drugs by high jet velocity through the skin without a needle. Among them, a new needle-free pyro-drive jet injector has a unique characteristic that utilizes gunpower as a mechanical driving force, in particular, bi-phasic pyrotechnics to provoke high jet velocity and consequently the wide dispersion of the injected DNA solution in the skin. A significant amount of evidence has revealed that it is highly effective as a vaccinating tool to induce potent protective cellular and humoral immunity against cancers and infectious diseases. This is presumably explained by the fact that shear stress generated by the high jet velocity facilitates the uptake of DNA in the cells and, consequently, its protein expression. The shear stress also possibly elicits danger signals which, together with the plasmid DNA, subsequently induces the activation of innate immunity including dendritic cell maturation, leading to the establishment of adaptive immunity. This review summarizes the recent advances in needle-free jet injectors to augment the cellular and humoral immunity by intradermal injection and the possible mechanism of action.
Collapse
Affiliation(s)
- Jukito Sonoda
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Izuru Mizoguchi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Shinya Inoue
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Aruma Watanabe
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Ami Sekine
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Miu Yamagishi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Satomi Miyakawa
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Natsuki Yamaguchi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Eri Horio
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Yasuhiro Katahira
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Hideaki Hasegawa
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Takashi Hasegawa
- Department of Device Application for Molecular Therapeutics, Graduate School of Medicine, Osaka University, CoMIT 0603, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Kunihiko Yamashita
- Department of Device Application for Molecular Therapeutics, Graduate School of Medicine, Osaka University, CoMIT 0603, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Takayuki Yoshimoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| |
Collapse
|
44
|
Marascio N, Loria MT, Pavia G, Peronace C, Adams NJ, Campolo M, Divenuto F, Lamberti AG, Giancotti A, Barreca GS, Mazzitelli M, Trecarichi EM, Torti C, Perandin F, Bisoffi Z, Quirino A, Matera G. Evaluation of IL-35, as a Possible Biomarker for Follow-Up after Therapy, in Chronic Human Schistosoma Infection. Vaccines (Basel) 2023; 11:vaccines11050995. [PMID: 37243099 DOI: 10.3390/vaccines11050995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The host response to helminth infections is characterized by systemic and tissue-related immune responses that play a crucial role in pathological diseases. Recently, experimental studies have highlighted the role of regulatory T (Tregs) and B (Bregs) cells with secreted cytokines as important markers in anti-schistosomiasis immunity. We investigated the serical levels of five cytokines (TNFα, IFN-γ, IL-4, IL-10 and IL-35) in pre- and post-treatment samples from chronic Schistosoma infected patients to identify potential serological markers during follow-up therapy. Interestingly, we highlighted an increased serum level of IL-35 in the pre-therapy samples (median 439 pg/mL for Schistosoma haematobium and 100.5 pg/mL for Schistsoma mansoni infected patients) compared to a control group (median 62 pg/mL and 58 pg/mL, respectively, p ≤ 0.05), and a significantly lower concentration in post-therapy samples (181 pg/mL for S. haematobium and 49.5 pg/mL for S. mansoni infected patients, p ≤ 0.05). The present study suggests the possible role of IL-35 as a novel serological biomarker in the evaluation of Schistosoma therapy follow-up.
Collapse
Affiliation(s)
- Nadia Marascio
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Maria Teresa Loria
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Grazia Pavia
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Cinzia Peronace
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Neill James Adams
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Morena Campolo
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Francesca Divenuto
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Angelo Giuseppe Lamberti
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Aida Giancotti
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Giorgio Settimo Barreca
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Maria Mazzitelli
- Infectious and Tropical Diseases Unit, Padua University Hospital, 35128 Padua, Italy
| | - Enrico Maria Trecarichi
- Infectious and Tropical Diseases Unit, Department of Medical and Surgical Sciences, "Magna Graecia" University-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Carlo Torti
- Infectious and Tropical Diseases Unit, Department of Medical and Surgical Sciences, "Magna Graecia" University-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Francesca Perandin
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, 37024 Verona, Italy
| | - Zeno Bisoffi
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, 37024 Verona, Italy
| | - Angela Quirino
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Giovanni Matera
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| |
Collapse
|
45
|
Li H, Xing H. Interleukin-35 Enhances Regulatory T Cell Function by Potentially Suppressing Their Transdifferentiation into a T Helper 17-Like Phenotype in Kawasaki Disease. Immunol Invest 2023:1-16. [PMID: 37052682 DOI: 10.1080/08820139.2023.2201283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Interleukin-35 (IL-35) modulates immune cell activity in inflammation and autoimmune disorders. However, its specific effects on regulatory T cells (Tregs) in Kawasaki disease remain ambiguous. We enrolled 37 patients with Kawasaki disease and 20 healthy controls in this study. The percentages of CD4+CD25+CD127dim/- Tregs and CD4+IL-17A+ T helper 17 (Th17) cells were determined via flow cytometry. Tregs were enriched and stimulated by recombinant IL-35. Immunosuppressive activity of Tregs was via co-culture with autologous CD4+CD25- T cells. Purified Tregs were cultured for Th17 polarization, and the influence of IL-35 on Tregs transdifferentiation into a Th17-like phenotype was determined. The percentage of Tregs was elevated in patients with Kawasaki disease and positively correlated with C-reactive protein levels. There was no significant difference in the percentage of Th17 cells between the two groups. IL-35 stimulation increased the percentage of Tregs in both groups, but decreased the percentage of Tregs Th17 cells in affected patients. IL-35 enhanced the immunosuppressive activity of Tregs in both groups, resulting in decreased cellular proliferation and increased IL-35 subunit mRNA relative levels in co-culture system. IL-35 did not affect the immune checkpoint molecule expression in Tregs, but inhibited the transdifferentiation of Tregs into a Th17-like phenotype in affected patients, indicating by the down-regulations of C-C motif chemokine receptor-4/6 expression, retinoid-related orphan nuclear receptor γt mRNA levels, and IL-17 secretion. IL-35 contributes to the immunosuppressive function of Tregs by inhibiting the cellular proliferation and transdifferentiation of Tregs into a Th17-like phenotype, which may be a protective mechanism against Kawasaki disease.
Collapse
Affiliation(s)
- Hua Li
- Department of Disease Prevention and Control, Xi'an Children's Hospital, The Children's Hospital Affiliated to Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Haijian Xing
- Department of General Internal Medicine, Xi'an Children's Hospital, The Children's Hospital Affiliated to Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
46
|
Interleukin-35 -producing B cells rescues inflammatory bowel disease in a mouse model via STAT3 phosphorylation and intestinal microbiota modification. Cell Death Discov 2023; 9:67. [PMID: 36797242 PMCID: PMC9935866 DOI: 10.1038/s41420-023-01366-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Interleukin-35 (IL-35)-producing B cells (IL-35+B cells) play an important role in diseases, and the expansion of IL-35+ immune cells have been observed in inflammatory bowel disease (IBD). However, how IL-35+B cells function and the manner in which they perform their roles remain unclear. In this study, human samples and animal models were used to confirm the expansion of IL-35+B cells during IBD. In addition, by using il12a-/- and ebi3-/- mice, we demonstrated that the regulatory role of B cells in IBD depends on IL-35. Mechanically, IL-35+B cells can promote its own expansion through endocrine actions and depend on the transcription factor signal transducer and activator of transcription 3. Interestingly, we found that the diversity of intestinal microbes and expression of microbial metabolites decreased during IBD. IL-35+B cells promote the high expression of indoleacetic acid (IAA), and exogenous metabolite supplementation with IAA can further promote the expansion of IL-35+B cells and rescues the disease. This study provides a new concept for the regulatory model of B cells and a new approach for the treatment of IBD.
Collapse
|
47
|
Liang H, Shi H, Li Y, Wang D, Zhang Y. Mechanism of Aspirin oxidative stress regulating interleukin-induced apoptosis in nucleus pulposus cells in a rat model of intervertebral disc degeneration. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:124. [PMID: 36819489 PMCID: PMC9929745 DOI: 10.21037/atm-22-5700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/11/2023] [Indexed: 01/31/2023]
Abstract
Background Intervertebral disc degeneration (IDD) is an important cause of low back pain. Increase of reactive oxygen species (ROS), overexpression of inflammatory factors, and loss of extracellular matrix are important factors in the pathological changes of IDD. The present study aimed to investigate the mechanism of action of Aspirin regulating oxidative stress in IDD, so as to propose new treatment. Methods Nucleus pulposus cells (NPCs) were isolated from the caudal intervertebral discs of Sprague Dawley (SD) rats under sterile conditions. The expression of ROS and inflammatory factors was detected sequentially, and the degree of degeneration of nucleus pulposus cells was observed by real-time fluorescence quantitative polymerase chain reaction (PCR) and cell immunofluorescence staining. In vivo, the caudal disc puncture model was used to induce degeneration, and a local injection of 10 or 100 µg/mL Aspirin was performed. The rats were sacrificed 1 week later, and the disc specimens of the tail vertebrae were collected for imaging, histomorphology, and immunohistochemical analysis. Results In vitro experiments showed that lipopolysaccharide (LPS) could significantly induce oxidative stress in NPCs and stimulate NPCs to secrete a large amount of ROS and inflammatory factors, which eventually leads to the reduction of collagen type II and polyglycoprotein gene expression in NPCs and the high expression of matrix metalloproteinase (MMP). Consequently, NPCs degeneration occurs. Conclusions Our results clarified the important role of oxidative stress in IDD and proved that LPS can be used as a drug to alleviate oxidative stress and intervene in the IDD process.
Collapse
Affiliation(s)
- He Liang
- Department of Orthopedics, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Hongyang Shi
- Second Department of Spine Surgery, Changzheng Hospital, Shanghai, China
| | - Yang Li
- Department of Orthopedics, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Deguo Wang
- Department of Orthopedics, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Yajie Zhang
- College of pharmacy, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
48
|
Saad H, Soliman HA, Mahmoud B, Moneim AA, Zaky MY. The Pathogenic Role of Oxidative Stress, Cytokine Expression, and Impaired Hematological Indices in Diabetic Cardiovascular Diseases. Inflammation 2023; 46:146-160. [PMID: 35997998 PMCID: PMC9971070 DOI: 10.1007/s10753-022-01718-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/28/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022]
Abstract
A simultaneous increase in the prevalence of diabetes mellitus (DM), a risk factor for cardiovascular diseases (CVDs), has contributed to the escalation of CVD related mortalities. To date, oxidative stress and inflammation are increasingly recognized as significant drivers of cardiovascular complications in patients with diabetes. Therefore, this study aims to explore the correlation between oxidative stress, inflammation, and hematological indices in diabetic patients with CVDs. Patients were allocated into five groups: healthy controls; nondiabetic patients with myocardial infarction; diabetic patients with myocardial infarction; nondiabetic patients with heart failure; and diabetic patients with heart failure. The results revealed that the malondialdehyde levels were increased; whereas superoxide dismutase enzyme activities were markedly reduced in all CVD groups compared with those of healthy controls. Although the mRNA expression levels of interleukin (IL)-6, IL-18, and IL-38 were significantly increased, those of the anti-inflammatory cytokine, IL-35, have been reduced in all CVD groups compared with healthy controls. Regarding hematological indices, hematocrit, red blood cell distribution width, mean platelet (PLT) volume, plateletcrit, PLT distribution width, leukocyte count, and PLT-to-lymphocyte and neutrophil-to-lymphocyte ratios were markedly increased in the diabetic and nondiabetic CVD groups compared with those of the healthy controls. Oxidative stress and cytokine biomarkers may play a significant role in the complications of diabetic cardiomyopathy. Moreover, hematological indices are particularly sensitive to systemic inflammatory changes and are novel markers for the early detection of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Howaida Saad
- Department of Biochemistry Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Hanan A. Soliman
- Department of Biochemistry Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Basant Mahmoud
- Department of Biochemistry Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Adel Abdel Moneim
- Molecular Physiology Division, Faculty of Science, Beni-Suef University, Salah Salem St, Beni-Suef, 62511 Egypt
| | - Mohamed Y. Zaky
- Molecular Physiology Division, Faculty of Science, Beni-Suef University, Salah Salem St, Beni-Suef, 62511 Egypt
| |
Collapse
|
49
|
Feng J, Wu Y. Interleukin-35 ameliorates cardiovascular disease by suppressing inflammatory responses and regulating immune homeostasis. Int Immunopharmacol 2022; 110:108938. [PMID: 35759811 DOI: 10.1016/j.intimp.2022.108938] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/05/2022] [Accepted: 06/07/2022] [Indexed: 12/14/2022]
Abstract
The immune response is of great significance in the initiation and progression of a diversity of cardiovascular diseases involving pro-and anti-inflammatory cytokines. Interleukin-35 (IL-35), a cytokine of the interleukin-12 family, is a novel anti-inflammation and immunosuppressive cytokine, maintaining inflammatory suppression and regulating immune homeostasis. The role of IL-35 in cardiovascular diseases (CVDs) has aroused enthusiastic attention, a diversity of experimental or clinical evidence has indicated that IL-35 potentially has a pivot role in protecting against cardiovascular diseases, especially atherosclerosis and myocarditis. In this review, we initiate an overview of the relationship between Interleukin-35 and cardiovascular diseases, including atherosclerosis, acute coronary syndrome, pulmonary hypertension, abdominal aortic aneurysm, heart failure, myocardial ischemia-reperfusion, aortic dissection and myocarditis. Although the specific molecular mechanisms entailing the protective effects of IL-35 remain an unsolved issue, targeted therapies with IL-35 might provide a promising and effective solution to prevent and cure cardiovascular diseases.
Collapse
Affiliation(s)
- Jie Feng
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
50
|
Hildenbrand K, Aschenbrenner I, Franke FC, Devergne O, Feige MJ. Biogenesis and engineering of interleukin 12 family cytokines. Trends Biochem Sci 2022; 47:936-949. [PMID: 35691784 DOI: 10.1016/j.tibs.2022.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/04/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023]
Abstract
Interleukin 12 (IL-12) family cytokines are secreted proteins that regulate immune responses. Each family member is a heterodimer and nature uses shared building blocks to assemble the functionally distinct IL-12 cytokines. In recent years we have gained insights into the molecular principles and cellular regulation of IL-12 family biogenesis. For each of the family members, generally one subunit depends on its partner to acquire its native structure and be secreted from immune cells. If unpaired, molecular chaperones retain these subunits in cells. This allows cells to regulate and control secretion of the highly potent IL-12 family cytokines. Molecular insights gained into IL-12 family biogenesis, structure, and function now allow us to engineer IL-12 family cytokines to develop novel immunotherapeutic approaches.
Collapse
Affiliation(s)
- Karen Hildenbrand
- Department of Chemistry, Technical University of Munich, 85748 Garching, Germany
| | - Isabel Aschenbrenner
- Department of Chemistry, Technical University of Munich, 85748 Garching, Germany
| | - Fabian C Franke
- Department of Chemistry, Technical University of Munich, 85748 Garching, Germany
| | - Odile Devergne
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), 75 013 Paris, France.
| | - Matthias J Feige
- Department of Chemistry, Technical University of Munich, 85748 Garching, Germany.
| |
Collapse
|