1
|
Yang Y, Wang L, Yu L, Chang C, Zhang H, Hu L, Liu J, Zhang Y, Han H, Zhang H, Zhou Y, Wang J. Monocytes Expressing IL-36G Play a Crucial Role in Atopic Dermatitis. J Cell Mol Med 2025; 29:e70503. [PMID: 40159643 PMCID: PMC11955416 DOI: 10.1111/jcmm.70503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
Atopic dermatitis (ad) is a chronic inflammatory skin disease, with recent studies indicating that immune cells, such as monocytes and inflammatory cytokines, play a crucial role. By retrieving datasets from public databases and analysing immune cell infiltration in lesional skin using CIBERSORT, we found that monocytes and M2 macrophages were significantly upregulated in atopic dermatitis. Differentially expressed gene (DEG) functional enrichment analysis revealed that cytokine-cytokine receptor interaction was the most significantly enriched pathway. Further analysis of cytokines and their receptors, along with their correlation with infiltrating immune cells, identified IL36G-expressing monocytes as a key target in atopic dermatitis. We compared immune cell infiltration and cytokine-related targets in similar inflammatory skin diseases, such as psoriasis and urticaria, to evaluate similarities and differences among these three skin conditions. The analysis revealed that IL36G-expressing monocytes were also highly expressed in psoriasis but did not play a pivotal role in urticaria. Finally, we used molecular docking to predict and validate drugs targeting IL36G. Our study highlights IL36G-expressing monocytes as a common key target in atopic dermatitis and psoriasis, offering novel insights and therapeutic strategies for these related diseases.
Collapse
Affiliation(s)
- Yitao Yang
- School of MedicineShanghai UniversityShanghaiChina
| | - Lei Wang
- Hubei Shizhen LaboratoryHubei University of Chinese MedicineWuhanChina
| | - Longmei Yu
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Chenxi Chang
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Honglei Zhang
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Linhan Hu
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Juntong Liu
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Yihang Zhang
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Hui Han
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Haiyun Zhang
- School of MedicineShanghai UniversityShanghaiChina
| | - Yumei Zhou
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Ji Wang
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| |
Collapse
|
2
|
Ahmad F, Alam MA, Ansari AW, Jochebeth A, Leo R, Al-Abdulla MN, Al-Khawaga S, AlHammadi A, Al-Malki A, Al Naama K, Ahmad A, Buddenkotte J, Steinhoff M. Emerging Role of the IL-36/IL-36R Axis in Multiple Inflammatory Skin Diseases. J Invest Dermatol 2024; 144:206-224. [PMID: 38189700 DOI: 10.1016/j.jid.2023.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/26/2023] [Accepted: 11/14/2023] [Indexed: 01/09/2024]
Abstract
IL-36 is a most recent member of the IL-1 cytokine family, primarily expressed at barrier sites of the body such as the skin, lungs, and intestine. It plays a vital role in inflammation and is implicated in the development of various cutaneous; intestinal; and pulmonary disorders, including psoriasis, inflammatory bowel disease, and chronic obstructive pulmonary disease. IL-36 comprises 4 isoforms: the proinflammatory IL-36α, IL-36β, and IL-36γ and the anti-inflammatory IL-36R antagonist. An imbalance between proinflammatory and anti-inflammatory IL-36 isoforms can contribute to the inflammatory fate of cells and tissues. IL-36 cytokines signal through an IL-36R heterodimer mediating their function through canonical signaling cacade, including the NF-B pathway. Prominent for its role in psoriasis, IL-36 has recently been associated with disease mechanisms in atopic dermatitis, hidradenitis suppurativa, neutrophilic dermatoses, autoimmune blistering disease, and Netherton syndrome. The major cutaneous source of IL-36 cytokines is keratinocytes, pointing to its role in the communication between the epidermis, innate (neutrophils, dendritic cells) immune system, and adaptive (T helper [Th]1 cells, Th17) immune system. Thus, cutaneous IL-36 signaling is crucial for the immunopathological outcome of various skin diseases. Consequently, the IL-36/IL-36R axis has recently been recognized as a promising drug target for the treatment of inflammatory disorders beyond psoriasis. This review summarizes the current update on IL-36 cytokines in inflammatory skin diseases.
Collapse
Affiliation(s)
- Fareed Ahmad
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Majid Ali Alam
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Abdul Wahid Ansari
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Anh Jochebeth
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Rari Leo
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | | | - Sara Al-Khawaga
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Ayda AlHammadi
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Aysha Al-Malki
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Khalifa Al Naama
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Aamir Ahmad
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Jörg Buddenkotte
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| | - Martin Steinhoff
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Medical School, Qatar University, Doha, Qatar; Weill Cornell Medicine, Weill Cornell University, New York, New York, USA; Weill Cornell Medicine-Qatar, Doha, Qatar.
| |
Collapse
|
3
|
Andoh A, Nishida A. Pro- and anti-inflammatory roles of interleukin (IL)-33, IL-36, and IL-38 in inflammatory bowel disease. J Gastroenterol 2023; 58:69-78. [PMID: 36376594 DOI: 10.1007/s00535-022-01936-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/29/2022] [Indexed: 11/16/2022]
Abstract
Interleukin-33 (IL-33), IL-36, and IL-38 are members of the IL-1 cytokine family. The expression of each cytokine has been reported to be increased in the inflamed mucosa of patients with inflammatory bowel disease (IBD). IL-33 and IL-36 have been studied for pro- and anti-inflammatory functions, and IL-38 has been characterized as an anti-inflammatory cytokine by antagonizing the IL-36 receptor (IL-36R). IL-33 is a nuclear cytokine constitutively expressed by certain cell types such as epithelial, endothelial, and fibroblast-like cells and released on necrotic cell death. IL-33 mainly induces type 2 immune response through its receptor suppression tumorigenicity 2 (ST2) from Th2 cells and type 2 innate lymphoid cells (ILC2s), but also by stimulating Th1 cells, regulatory T cells, and CD8+ T cells. IL-36 cytokines consist of three agonists: IL-36α, IL-36β, and IL-36γ, and two receptor antagonists: IL-36R antagonist (IL-36Ra) and IL-38. All IL-36 cytokines bind to the IL-36R complex and exert various functions through NF-κB and mitogen-activated protein kinase (MAPK) pathways in inflammatory settings. IL-33 and IL-36 also play a crucial role in intestinal fibrosis characteristic manifestation of CD. In this review, we focused on the current understanding of the pro- and anti-inflammatory roles of IL-33, IL-36, and IL38 in experimental colitis and IBD patients.
Collapse
Affiliation(s)
- Akira Andoh
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu, Shiga, 520-2192, Japan.
| | - Atsushi Nishida
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|
4
|
Purewal JS, Doshi GM. Deciphering the Function of New Therapeutic Targets and Prospective Biomarkers in the Management of Psoriasis. Curr Drug Targets 2023; 24:1224-1238. [PMID: 38037998 DOI: 10.2174/0113894501277656231128060242] [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: 08/17/2023] [Revised: 10/29/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023]
Abstract
Psoriasis is an immune-mediated skin condition affecting people worldwide, presenting at any age, and leading to a substantial burden physically and mentally. The innate and adaptive immune systems interact intricately with the pathomechanisms that underlie disease. T cells can interact with keratinocytes, macrophages, and dendritic cells through the cytokines they secrete. According to recent research, psoriasis flare-ups can cause systemic inflammation and various other co-morbidities, including depression, psoriatic arthritis, and cardio-metabolic syndrome. Additionally, several auto-inflammatory and auto-immune illnesses may be linked to psoriasis. Although psoriasis has no proven treatment, care must strive by treating patients as soon as the disease surfaces, finding and preventing concurrent multimorbidity, recognising and reducing bodily and psychological distress, requiring behavioural modifications, and treating each patient individually. Biomarkers are traits that are assessed at any time along the clinical continuum, from the early stages of a disease through the beginning of treatment (the foundation of precision medicine) to the late stages of treatment (outcomes and endpoints). Systemic therapies that are frequently used to treat psoriasis provide a variety of outcomes. Targeted therapy selection, better patient outcomes, and more cost-effective healthcare would be made possible by biomarkers that reliably predict effectiveness and safety. This review is an attempt to understand the role of Antimicrobial peptides (AMP), Interleukin-38 (IL-38), autophagy 5 (ATG5) protein and squamous cell carcinoma antigen (SCCA) as biomarkers of psoriasis.
Collapse
Affiliation(s)
- Japneet Singh Purewal
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, India
| | - Gaurav Mahesh Doshi
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, India
| |
Collapse
|
5
|
Haghshenas MR, Zamir MR, Sadeghi M, Fattahi MJ, Mirshekari K, Ghaderi A. Clinical relevance and therapeutic potential of IL-38 in immune and non-immune-related disorders. Eur Cytokine Netw 2022; 33:54-69. [PMID: 37052152 PMCID: PMC10134710 DOI: 10.1684/ecn.2022.0480] [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] [Accepted: 07/30/2022] [Indexed: 04/14/2023]
Abstract
Interleukin-38 (IL-38) is the most recent member of the IL-1 family that acts as a natural inflammatory inhibitor by binding to cognate receptors, particularly the IL-36 receptor. In vitro, animal and human studies on autoimmune, metabolic, cardiovascular and allergic diseases, as well sepsis and respiratory viral infections, have shown that IL-38 exerts an anti-inflammatory activity by modulating the generation and function of inflammatory cytokines (e.g. IL-6, IL-8, IL-17 and IL-36) and regulating dendritic cells, M2 macrophages and regulatory T cells (Tregs). Accordingly, IL-38 may possess therapeutic potential for these types of diseases. IL-38 down-regulates CCR3+ eosinophil cells, CRTH2+ Th2 cells, Th17 cells, and innate lymphoid type 2 cells (ILC2), but up-regulates Tregs, and this has influenced the design of immunotherapeutic strategies based on regulatory cells/cytokines for allergic asthma in future studies. In auto-inflammatory diseases, IL-38 alleviates skin inflammation by regulating γδ T cells and limiting the production of IL-17. Due to its ability to suppress IL-1β, IL-6 and IL-36, this cytokine could reduce COVID-19 severity, and might be employed as a therapeutic tool. IL-38 may also influence host immunity and/or the components of the cancer microenvironment, and has been shown to improve the outcome of colorectal cancer, and may participate in tumour progression in lung cancer possibly by modulating CD8 tumour infiltrating T cells and PD-L1 expression. In this review, we first briefly present the biological and immunological functions of IL-38, and then discuss the important roles of IL-38 in various types of diseases, and finally highlight its use in therapeutic strategies.
Collapse
Affiliation(s)
- Mohammad Reza Haghshenas
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mina Roshan Zamir
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboubeh Sadeghi
- 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
| | - Kimia Mirshekari
- Shiraz Institute for Cancer Research, 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
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
6
|
Poudel M, Bhattarai PY, Shrestha P, Choi HS. Regulation of Interleukin-36γ/IL-36R Signaling Axis by PIN1 in Epithelial Cell Transformation and Breast Tumorigenesis. Cancers (Basel) 2022; 14:cancers14153654. [PMID: 35954317 PMCID: PMC9367291 DOI: 10.3390/cancers14153654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/15/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Members of the interleukin (IL)-1 cytokine family exhibit dual functions in the regulation of inflammation and cancer. Recent studies have shown the critical role of IL-36γ, the newly identified IL-1 family member, in the regulation of cellular processes implicated in the progression of cancer. Therefore, the underlying mechanism of IL-36γ in tumor development is of considerable interest. Here, we identified the pivotal role of IL-36γ in the proliferation of breast cancer cells. Consistently, IL-36γ was found to promote epithelial cell transformation via the activation of c-Fos, c-Jun, and AP-1 transcription factors, followed by the IL36R-mediated MEK/ERK and JNK/c-Jun cascades. Furthermore, our findings demonstrate the critical role of PIN1 in the regulation of IL-36γ-induced mammary gland tumorigenesis. Abstract Given the increasing recognition of the relationship between IL-1 cytokines, inflammation, and cancer, the significance of distinct members of the IL-1 cytokine family in the etiology of cancer has been widely researched. In the present study, we investigated the underlying mechanism of the IL-36γ/IL-36R axis during breast cancer progression, which has not yet been elucidated. Initially, we determined the effects of IL-36γ on the proliferation and epithelial cell transformation of JB6 Cl41 mouse epidermal and MCF7 human breast cancer cells using BrdU incorporation and anchorage-independent growth assays. We found that treatment with IL-36γ increased the proliferation and colony formation of JB6 Cl41 and MCF7 cells. Analysis of the mechanism underlying the neoplastic cell transformation revealed that IL-36γ induced IL-36R-mediated phosphorylation of MEK1/2, ERK1/2, JNK1/2, and c-Jun, resulting in increased c-Fos, c-Jun, and AP-1 activities in JB6 Cl41 and MCF7 cells. Furthermore, the IL-36γ-induced tumorigenic capacity of MCF7 cells was considerably enhanced by PIN1, following MEK/ERK and JNK/c-Jun signaling. Interestingly, blocking PIN1 activity using juglone suppressed the IL-36γ-induced increase in the anchorage-independent growth of 4T1 metastatic mouse breast cancer cells. Finally, in a syngeneic mouse model, IL-36γ-induced tumor growth in the breast mammary gland was significantly inhibited following PIN1 knockout.
Collapse
Affiliation(s)
| | | | | | - Hong Seok Choi
- Correspondence: ; Tel.: +82-622306379; Fax: +82-622225414
| |
Collapse
|
7
|
Le N, Luk I, Chisanga D, Shi W, Pang L, Scholz G, Mariadason J, Ernst M, Huynh J. IL-36G promotes cancer-cell intrinsic hallmarks in human gastric cancer cells. Cytokine 2022; 155:155887. [PMID: 35512531 DOI: 10.1016/j.cyto.2022.155887] [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: 10/28/2021] [Revised: 03/24/2022] [Accepted: 04/07/2022] [Indexed: 11/28/2022]
Abstract
Interleukin-36 gamma (IL-36G) is a member of the IL-36 subfamily of cytokines and acts as a potent driver of inflammation. IL-36G has been extensively characterized in the pathogenesis of psoriasis and has been recently described to play roles in wound healing particularly in the gastrointestinal tract. However, the effects of IL-36G during cancer development including gastric cancer remain unexplored. Here, we show that IL-36G induced ERK1/2 activation in AGS, MKN1 and MKN45 human gastric cancer cell lines. Moreover, IL-36G induced colony formation, migration and invasion of these gastric cancer cell lines that was inhibited by the natural antagonist, IL-36 receptor antagonist (RA). Interrogation of TCGA stomach adenocarcinoma patient datasets revealed highly elevated IL-36G gene expression in human gastric cancer compared to normal tissue independent of tumor stage, and high IL-36G expression corresponded with poorer patient survival. Collectively, our results indicate for the first time that IL-36G supports a neoplastic phenotype in human gastric cancer cells.
Collapse
Affiliation(s)
- Ngan Le
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia
| | - Ian Luk
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia
| | - David Chisanga
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia; School of Computing and Information Systems, The University of Melbourne, Parkville, VIC, Australia
| | - Wei Shi
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia; School of Computing and Information Systems, The University of Melbourne, Parkville, VIC, Australia
| | - Lokman Pang
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia
| | - Glen Scholz
- Melbourne Dental School, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, VIC, Australia
| | - John Mariadason
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia
| | - Jennifer Huynh
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, VIC, Australia
| |
Collapse
|
8
|
Sun R, Gao DS, Shoush J, Lu B. The IL-1 family in tumorigenesis and antitumor immunity. Semin Cancer Biol 2022; 86:280-295. [DOI: 10.1016/j.semcancer.2022.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/24/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022]
|
9
|
Frühbeck G, Gómez-Ambrosi J, Ramírez B, Mentxaka A, Rodríguez A, Becerril S, Reina G, Valentí V, Moncada R, Silva C, Catalán V. Increased Levels of Interleukin-36 in Obesity and Type 2 Diabetes Fuel Adipose Tissue Inflammation by Inducing Its Own Expression and Release by Adipocytes and Macrophages. Front Immunol 2022; 13:832185. [PMID: 35222417 PMCID: PMC8863603 DOI: 10.3389/fimmu.2022.832185] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/20/2022] [Indexed: 02/03/2023] Open
Abstract
Interleukin (IL)-36 is a recently described cytokine with well-known functions in the regulation of multiple inflammatory diseases. Since no data exists on how this cytokine regulates adipose tissue (AT) homeostasis, we aimed to explore the function of a specific isoform, IL-36γ, an agonist, in human obesity and obesity-associated type 2 diabetes as well as in AT inflammation and fibrosis. Plasma IL-36γ was measured in 91 participants in a case-control study and the effect of weight loss was evaluated in 31 patients with severe obesity undergoing bariatric surgery. Gene expression levels of IL36G and its receptor were analyzed in relevant human metabolic tissues. The effect of inflammatory factors and IL-36γ was determined in vitro in human adipocytes and macrophages. We found, for the first time, that the increased (P<0.05) circulating levels of IL-36γ in patients with obesity decreased (P<0.001) after weight and fat loss achieved by Roux-en-Y gastric bypass and that gene expression levels of IL36G were upregulated in the visceral AT (P<0.05) and in the peripheral blood mononuclear cells (P<0.01) from patients with obesity. We also demonstrated increased (P<0.05) expression levels of Il36g in the epididymal AT from diet-induced obese mice. IL36G was significantly enhanced (P<0.001) by LPS in human adipocytes and monocyte-derived macrophages, while no changes were found after the incubation with anti-inflammatory cytokines. The addition of IL-36γ for 24 h strongly induced (P<0.01) its own expression as well as key inflammatory and chemoattractant factors with no changes in genes associated with fibrosis. Furthermore, adipocyte-conditioned media obtained from patients with obesity increased (P<0.01) the release of IL-36γ and the expression (P<0.05) of cathepsin G (CTSG) in monocyte-derived macrophages. These findings provide, for the first time, evidence about the properties of IL-36γ in the regulation of AT-chronic inflammation, emerging as a link between AT biology and the obesity-associated comorbidities.
Collapse
Affiliation(s)
- Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain,Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain,*Correspondence: Victoria Catalán, ; Gema Frühbeck,
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Amaia Mentxaka
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Gabriel Reina
- Department of Microbiology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victor Valentí
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Camilo Silva
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain,*Correspondence: Victoria Catalán, ; Gema Frühbeck,
| |
Collapse
|
10
|
Peñaloza HF, van der Geest R, Ybe JA, Standiford TJ, Lee JS. Interleukin-36 Cytokines in Infectious and Non-Infectious Lung Diseases. Front Immunol 2021; 12:754702. [PMID: 34887860 PMCID: PMC8651476 DOI: 10.3389/fimmu.2021.754702] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/14/2021] [Indexed: 12/26/2022] Open
Abstract
The IL-36 family of cytokines were identified in the early 2000’s as a new subfamily of the IL-1 cytokine family, and since then, the role of IL-36 cytokines during various inflammatory processes has been characterized. While most of the research has focused on the role of these cytokines in autoimmune skin diseases such as psoriasis and dermatitis, recent studies have also shown the importance of IL-36 cytokines in the lung inflammatory response during infectious and non-infectious diseases. In this review, we discuss the biology of IL-36 cytokines in terms of how they are produced and activated, as well as their effects on myeloid and lymphoid cells during inflammation. We also discuss the role of these cytokines during lung infectious diseases caused by bacteria and influenza virus, as well as other inflammatory conditions in the lungs such as allergic asthma, lung fibrosis, chronic obstructive pulmonary disease, cystic fibrosis and cancer. Finally, we discuss the current therapeutic advances that target the IL-36 pathway and the possibility to extend these tools to treat lung inflammatory diseases.
Collapse
Affiliation(s)
- Hernán F Peñaloza
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rick van der Geest
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joel A Ybe
- Department of Environmental and Occupational Health, School of Public Health, Indiana University, Bloomington, IN, United States
| | - Theodore J Standiford
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Janet S Lee
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
11
|
SnapshotDx Quiz: August 2021. J Invest Dermatol 2021. [PMID: 34303470 DOI: 10.1016/j.jid.2021.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
12
|
Kang W, Feng Z, Luo J, He Z, Liu J, Wu J, Rong P. Tertiary Lymphoid Structures in Cancer: The Double-Edged Sword Role in Antitumor Immunity and Potential Therapeutic Induction Strategies. Front Immunol 2021; 12:689270. [PMID: 34394083 PMCID: PMC8358404 DOI: 10.3389/fimmu.2021.689270] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/05/2021] [Indexed: 12/14/2022] Open
Abstract
The complex tumor microenvironment (TME) plays a vital role in cancer development and dramatically determines the efficacy of immunotherapy. Tertiary lymphoid structures (TLSs) within the TME are well recognized and consist of T cell-rich areas containing dendritic cells (DCs) and B cell-rich areas containing germinal centers (GCs). Accumulating research has indicated that there is a close association between tumor-associated TLSs and favorable clinical outcomes in most types of cancers, though a minority of studies have reported an association between TLSs and a poor prognosis. Overall, the double-edged sword role of TLSs in the TME and potential mechanisms need to be further investigated, which will provide novel therapeutic perspectives for antitumor immunoregulation. In this review, we focus on discussing the main functions of TLSs in the TME and recent advances in the therapeutic manipulation of TLSs through multiple strategies to enhance local antitumor immunity.
Collapse
Affiliation(s)
- Wendi Kang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhichao Feng
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Molecular Imaging Research Center, Central South University, Changsha, China
| | - Jianwei Luo
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhenhu He
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jun Liu
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jianzhen Wu
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Molecular Imaging Research Center, Central South University, Changsha, China
| |
Collapse
|
13
|
Cytokine/Chemokine Release Patterns and Transcriptomic Profiles of LPS/IFNγ-Activated Human Macrophages Differentiated with Heat-Killed Mycobacterium obuense, M-CSF, or GM-CSF. Int J Mol Sci 2021; 22:ijms22137214. [PMID: 34281268 PMCID: PMC8268300 DOI: 10.3390/ijms22137214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 01/15/2023] Open
Abstract
Macrophages (Mφs) are instrumental regulators of the immune response whereby they acquire diverse functional phenotypes following their exposure to microenvironmental cues that govern their differentiation from monocytes and their activation. The complexity and diversity of the mycobacterial cell wall have empowered mycobacteria with potent immunomodulatory capacities. A heat-killed (HK) whole-cell preparation of Mycobacterium obuense (M. obuense) has shown promise as an adjunctive immunotherapeutic agent for the treatment of cancer. Moreover, HK M. obuense has been shown to trigger the differentiation of human monocytes into a monocyte-derived macrophage (MDM) type named Mob-MDM. However, the transcriptomic profile and functional properties of Mob-MDMs remain undefined during an activation state. Here, we characterized cytokine/chemokine release patterns and transcriptomic profiles of lipopolysaccharide (LPS)/interferon γ (IFNγ)-activated human MDMs that were differentiated with HK M. obuense (Mob-MDM(LPS/IFNγ)), macrophage colony-stimulating factor M-MDM(LPS/IFNγ)), or granulocyte/macrophage colony-stimulating factor (GM-MDM(LPS/IFNγ)). Mob-MDM(LPS/IFNγ) demonstrated a unique cytokine/chemokine release pattern (interleukin (IL)-10low, IL-12/23p40low, IL-23p19/p40low, chemokine (C-x-C) motif ligand (CXCL)9low) that was distinct from those of M-MDM(LPS/IFNγ) and GM-MDM(LPS/IFNγ). Furthermore, M-MDM(LPS/IFNγ) maintained IL-10 production at significantly higher levels compared to GM-MDM(LPS/IFNγ) and Mob-MDM(LPS/IFNγ) despite being activated with M1-Mφ-activating stimuli. Comparative RNA sequencing analysis pointed to a distinct transcriptome profile for Mob-MDM(LPS/IFNγ) relative to both M-MDM(LPS/IFNγ) and GM-MDM(LPS/IFNγ) that comprised 417 transcripts. Functional gene-set enrichment analysis revealed significant overrepresentation of signaling pathways and biological processes that were uniquely related to Mob-MDM(LPS/IFNγ). Our findings lay a foundation for the potential integration of HK M. obuense in specific cell-based immunotherapeutic modalities such as adoptive transfer of Mφs (Mob-MDM(LPS/IFNγ)) for cancer treatment.
Collapse
|
14
|
Ngo VL, Kuczma M, Maxim E, Denning TL. IL-36 cytokines and gut immunity. Immunology 2021; 163:145-154. [PMID: 33501638 DOI: 10.1111/imm.13310] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/19/2022] Open
Abstract
Interleukin 36 (IL-36) constitutes a group of cytokines that belong to the IL-1 superfamily. Emerging evidence has suggested a role of IL-36 in the pathogenesis of many inflammatory disorders. Intriguingly, in the gastrointestinal tract, IL-36 has a rather complex function. IL-36 receptor ligands are overexpressed in both animal colitis models and human IBD patients and may play both pathogenic and protective roles, depending on the context. IL-36 cytokines comprise three receptor agonists: IL-36α, IL-36β and IL-36γ, and two receptor antagonists: IL-36Ra and IL-38. All IL-36 receptor agonists bind to the IL-36R complex and exert pleiotropic effects during inflammatory settings. Here, we first briefly review the processing and secretion of IL-36 cytokines. We then focus on the current understanding of the immunology effects of IL-36 in gut immunity. In addition, we also discuss the ongoing trials that aim to blockage IL-36R signalling for treating chronic intestinal inflammation and present some unexplored questions regarding IL-36 research.
Collapse
Affiliation(s)
- Vu L Ngo
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Michal Kuczma
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Estera Maxim
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Timothy L Denning
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
15
|
Koss CK, Wohnhaas CT, Baker JR, Tilp C, Przibilla M, Lerner C, Frey S, Keck M, Williams CMM, Peter D, Ramanujam M, Fine J, Gantner F, Thomas M, Barnes PJ, Donnelly LE, El Kasmi KC. IL36 is a critical upstream amplifier of neutrophilic lung inflammation in mice. Commun Biol 2021; 4:172. [PMID: 33558616 PMCID: PMC7870940 DOI: 10.1038/s42003-021-01703-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/13/2021] [Indexed: 01/30/2023] Open
Abstract
IL-36, which belongs to the IL-1 superfamily, is increasingly linked to neutrophilic inflammation. Here, we combined in vivo and in vitro approaches using primary mouse and human cells, as well as, acute and chronic mouse models of lung inflammation to provide mechanistic insight into the intercellular signaling pathways and mechanisms through which IL-36 promotes lung inflammation. IL-36 receptor deficient mice exposed to cigarette smoke or cigarette smoke and H1N1 influenza virus had attenuated lung inflammation compared with wild-type controls. We identified neutrophils as a source of IL-36 and show that IL-36 is a key upstream amplifier of lung inflammation by promoting activation of neutrophils, macrophages and fibroblasts through cooperation with GM-CSF and the viral mimic poly(I:C). Our data implicate IL-36, independent of other IL-1 family members, as a key upstream amplifier of neutrophilic lung inflammation, providing a rationale for targeting IL-36 to improve treatment of a variety of neutrophilic lung diseases.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Cigarette Smoking
- Disease Models, Animal
- Female
- Fibroblasts/immunology
- Fibroblasts/metabolism
- Humans
- Influenza A Virus, H1N1 Subtype/pathogenicity
- Interleukin-1/genetics
- Interleukin-1/metabolism
- Lung/immunology
- Lung/metabolism
- Lung/virology
- Macrophage Activation
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/metabolism
- Male
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Neutrophil Activation
- Neutrophils/immunology
- Neutrophils/metabolism
- Neutrophils/virology
- Orthomyxoviridae Infections/genetics
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/metabolism
- Orthomyxoviridae Infections/virology
- Pneumonia, Viral/genetics
- Pneumonia, Viral/immunology
- Pneumonia, Viral/metabolism
- Pneumonia, Viral/virology
- Receptors, Interleukin-1/genetics
- Receptors, Interleukin-1/metabolism
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Carolin K Koss
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Christian T Wohnhaas
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Jonathan R Baker
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Cornelia Tilp
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | | | - Carmen Lerner
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Silvia Frey
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Martina Keck
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Cara M M Williams
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
- WRDM, Inflammation and Immunology Research Unit, Pfizer, Cambridge, MA, USA
| | - Daniel Peter
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Meera Ramanujam
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Jay Fine
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Florian Gantner
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Matthew Thomas
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Peter J Barnes
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Louise E Donnelly
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
16
|
Kovach MA, Che K, Brundin B, Andersson A, Asgeirsdottir H, Padra M, Lindén SK, Qvarfordt I, Newstead MW, Standiford TJ, Lindén A. IL-36 Cytokines Promote Inflammation in the Lungs of Long-Term Smokers. Am J Respir Cell Mol Biol 2021; 64:173-182. [PMID: 33105081 PMCID: PMC7874394 DOI: 10.1165/rcmb.2020-0035oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 10/26/2020] [Indexed: 12/21/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive inflammatory lung disease with high morbidity and mortality. The IL-36 family are proinflammatory cytokines that are known to shape innate immune responses, including those critical to bacterial pneumonia. The objective of this study was to determine whether IL-36 cytokines promote a proinflammatory milieu in the lungs of long-term smokers with and without COPD. Concentrations of IL-36 cytokines were measured in plasma and BAL fluid from subjects in a pilot study (n = 23) of long-term smokers with and without COPD in vivo and from a variety of lung cells (from 3-5 donors) stimulated with bacteria or cigarette smoke components in vitro. Pulmonary macrophages were stimulated with IL-36 cytokines in vitro, and chemokine and cytokine production was assessed. IL-36α and IL-36γ are produced to varying degrees in murine and human lung cells in response to bacterial stimuli and cigarette smoke components in vitro. Moreover, whereas IL-36γ production is upregulated early after cigarette smoke stimulation and wanes over time, IL-36α production requires a longer duration of exposure. IL-36α and IL-36γ are enhanced systemically and locally in long-term smokers with and without COPD, and local IL-36α concentrations display a positive correlation with declining ventilatory lung function and increasing proinflammatory cytokine concentrations. In vitro, IL-36α and IL-36γ induce proinflammatory chemokines and cytokines in a concentration-dependent fashion that requires IL-36R and MyD88. IL-36 cytokine production is altered in long-term smokers with and without COPD and contributes to shaping a proinflammatory milieu in the lungs.
Collapse
Affiliation(s)
- Melissa A. Kovach
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karlhans Che
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bettina Brundin
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anders Andersson
- Division of Respiratory Medicine and Allergology, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Helga Asgeirsdottir
- Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Médea Padra
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sara K. Lindén
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ingemar Qvarfordt
- Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Michael W. Newstead
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan; and
| | - Theodore J. Standiford
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan; and
| | - Anders Lindén
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
17
|
Analysis of Il36a induction by C/EBPβ via a half-CRE•C/EBP element in murine macrophages in dependence of its CpG methylation level. Genes Immun 2021; 22:313-321. [PMID: 34697411 PMCID: PMC8674125 DOI: 10.1038/s41435-021-00153-5] [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: 05/13/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 11/12/2022]
Abstract
Interleukin-36α is a novel member of the IL-1 cytokine family that is highly expressed in epithelial tissues and several myeloid-derived cell types after induction. The transcription factor (TF) C/EBPβ binds specifically to an essential half-CRE•C/EBP motif in the Il36a promoter to induce Il36a expression upon LPS stimulation. C/EBPs regulate gene expression by binding to recognition sequences that can contain 5'-cytosine-phosphate-guanine-3' dinucleotides (CpG), whose methylation can influence TF binding and gene expression. Herein we show that the half-CRE•C/EBP element in the Il36a promoter is differentially methylated in the murine RAW264.7 macrophage cell line and in primary murine macrophages. We demonstrate that C/EBPβ binding to the half-CRE•C/EBP element in the Il36a promoter following LPS stimulation is insensitive to CpG methylation and that methylation of the CpG in the half-CRE•C/EBP element does not alter LPS-induced Il36a promoter activity which correlated with similar Il36a mRNA copy numbers and pro-IL-36α protein amount in both cell types. Taken together, our data indicate that C/EBPβ binding to the half-CRE•C/EBP element and subsequent gene activation occurs independently of the CpG methylation status of the half-CRE•C/EBP motif and underlines the potential of C/EBPs to recognize methylated as well as unmethylated motifs.
Collapse
|
18
|
Satoh TK, Mellett M, Meier-Schiesser B, Fenini G, Otsuka A, Beer HD, Rordorf T, Maul JT, Hafner J, Navarini AA, Contassot E, French LE. IL-36γ drives skin toxicity induced by EGFR/MEK inhibition and commensal Cutibacterium acnes. J Clin Invest 2020; 130:1417-1430. [PMID: 31805013 PMCID: PMC7269569 DOI: 10.1172/jci128678] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 11/15/2019] [Indexed: 12/24/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) and MEK inhibitors (EGFRi/MEKi) are beneficial for the treatment of solid cancers but are frequently associated with severe therapy-limiting acneiform skin toxicities. The underlying molecular mechanisms are poorly understood. Using gene expression profiling we identified IL-36γ and IL-8 as candidate drivers of EGFRi/MEKi skin toxicity. We provide molecular and translational evidence that EGFRi/MEKi in concert with the skin commensal bacterium Cutibacterium acnes act synergistically to induce IL-36γ in keratinocytes and subsequently IL-8, leading to cutaneous neutrophilia. IL-36γ expression was the combined result of C. acnes–induced NF-κB activation and EGFRi/MEKi–mediated expression of the transcription factor Krüppel-like factor 4 (KLF4), due to the presence of both NF-κB and KLF4 binding sites in the human IL-36γ gene promoter. EGFRi/MEKi increased KLF4 expression by blockade of the EGFR/MEK/ERK pathway. These results provide an insight into understanding the pathological mechanism of the acneiform skin toxicities induced by EGFRi/MEKi and identify IL-36γ and the transcription factor KLF4 as potential therapeutic targets.
Collapse
Affiliation(s)
- Takashi K Satoh
- Department of Dermatology, University of Zürich, Zürich, Switzerland
| | - Mark Mellett
- Department of Dermatology, University of Zürich, Zürich, Switzerland
| | | | - Gabriele Fenini
- Department of Dermatology, University of Zürich, Zürich, Switzerland
| | - Atsushi Otsuka
- Department of Dermatology, Kyoto University, Kyoto, Japan
| | - Hans-Dietmar Beer
- Department of Dermatology, University of Zürich, Zürich, Switzerland.,Medical Faculty, University of Zürich, Zürich, Switzerland
| | - Tamara Rordorf
- Clinic for Oncology, University Hospital Zürich, Zürich, Switzerland
| | | | - Jürg Hafner
- Department of Dermatology, University of Zürich, Zürich, Switzerland.,Medical Faculty, University of Zürich, Zürich, Switzerland
| | - Alexander A Navarini
- Department of Dermatology, University of Zürich, Zürich, Switzerland.,Medical Faculty, University of Zürich, Zürich, Switzerland.,Department of Dermatology, University Hospital of Basel, Basel, Switzerland
| | - Emmanuel Contassot
- Department of Dermatology, University of Zürich, Zürich, Switzerland.,Medical Faculty, University of Zürich, Zürich, Switzerland
| | - Lars E French
- Department of Dermatology, University of Zürich, Zürich, Switzerland.,Medical Faculty, University of Zürich, Zürich, Switzerland.,Department of Dermatology and Allergology, Ludwig Maximilian University of Munich, Munich, Germany
| |
Collapse
|
19
|
Liu T, Wu H, Qi J, Qin C, Zhu Q. Seven immune-related genes prognostic power and correlation with tumor-infiltrating immune cells in hepatocellular carcinoma. Cancer Med 2020; 9:7440-7452. [PMID: 32815653 PMCID: PMC7571821 DOI: 10.1002/cam4.3406] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/24/2022] Open
Abstract
Background Given poor prognosis and the lack of efficient therapy for advanced hepatocellular carcinoma, immunotherapy has emerged as an increasingly important role. However, there were few reports on the correlation between immune‐related genes and HCC. The purpose of this study is to construct a novel immune‐related gene‐based prognostic signature for HCC and to explore the potential mechanisms. Methods We organized expression data of 374 HCC samples and 50 nontumor samples from TCGA database. A robust signature was constructed by Cox regression analysis based on the immune‐related genes, which were filtered by differential genes analysis and Cox regression analysis. Then, the correlation analysis between the signature and clinical characteristics was conducted. And the signature was validated in ICGC database. Furthermore, the relationships between immune cell infiltration and the signature were explored by bioinformatics analysis. Results Seven genes‐based model (Risk score = BIRC5 * 0.0238 + FOS * 0.0055 + DKK1 * 0.0085 + FGF13 * 0.3432 + IL11 * 0.0135 + IL17D * 0.0878 + SPP1 * 0.0003) was constructed eventually and it was proved to be an independent prognostic factor for HCC patients. The signature‐calculated risk scores were shown to be positively correlated with the infiltration of these five immune cells, including macrophages, neutrophils, CD8+T, dendritic, and B cells. And the results suggested that high amplication of BIRC5, FGF13, IL11, IL17D, and SPP1 were more likely correlated with immune cell infiltration. Finally, PPI network, TFs‐based regulatory network and gene enrichment plots were performed to show potential molecular mechanisms. Conclusion We construct a robust immune‐related gene‐based prognostic signature with seven genes and explore potential mechanisms about it, which may contribute to the immunotherapy research for HCC.
Collapse
Affiliation(s)
- Tiantian Liu
- Department of Gastroenterology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, Shandong, China
| | - Hao Wu
- Department of Gastroenterology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, Shandong, China
| | - Jianni Qi
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, Shandong, China.,Central Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chengyong Qin
- Department of Gastroenterology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, Shandong, China.,Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qiang Zhu
- Department of Gastroenterology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, Shandong, China.,Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
20
|
Goldstein JD, Bassoy EY, Caruso A, Palomo J, Rodriguez E, Lemeille S, Gabay C. IL-36 signaling in keratinocytes controls early IL-23 production in psoriasis-like dermatitis. Life Sci Alliance 2020; 3:e202000688. [PMID: 32345660 PMCID: PMC7190273 DOI: 10.26508/lsa.202000688] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 01/04/2023] Open
Abstract
IL-36R signaling plays an important role in the pathogenesis of psoriasis. We ought to assess the specific function of IL-36R in keratinocytes for the pathology of Aldara-induced psoriasis-like dermatitis. Il36r ΔK mice presenting deletion of IL-36R in keratinocytes were similarly resistant to Aldara-induced ear inflammation as Il36r -/- mice, but acanthosis was only prevented in Il36r -/- mice. FACS analysis revealed that IL-36R signaling in keratinocytes is mandatory for early neutrophil infiltration in Aldara-treated ears. RNASeq and qRT-PCR experiments demonstrated the crucial role of IL-36R signaling in keratinocytes for induction of IL-23, IL-17, and IL-22 at early time points. Taken together, our results demonstrate that IL-36R signaling in keratinocytes plays a major role in the induction of Aldara-induced psoriasis-like dermatitis by triggering early production of IL-23/IL-17/IL-22 cytokines and neutrophil infiltration.
Collapse
Affiliation(s)
- Jérémie D Goldstein
- Department of Pathology-Immunology, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Esen Y Bassoy
- Department of Pathology-Immunology, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Assunta Caruso
- Department of Pathology-Immunology, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Jennifer Palomo
- Department of Pathology-Immunology, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Emiliana Rodriguez
- Department of Pathology-Immunology, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Sylvain Lemeille
- Department of Pathology-Immunology, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Cem Gabay
- Department of Pathology-Immunology, University of Geneva Faculty of Medicine, Geneva, Switzerland
- Division of Rheumatology, Department of Medicine, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
21
|
Qu Q, Zhai Z, Xu J, Li S, Chen C, Lu B. IL36 Cooperates With Anti-CTLA-4 mAbs to Facilitate Antitumor Immune Responses. Front Immunol 2020; 11:634. [PMID: 32351508 PMCID: PMC7174717 DOI: 10.3389/fimmu.2020.00634] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 03/19/2020] [Indexed: 12/23/2022] Open
Abstract
Despite the great impact on long-term survival of some cancer patients, the immune checkpoint blockade (ICB) therapy is limited by its low response rates for most cancers. There is a pressing need for novel combination immunotherapies that overcome the resistance to current ICB therapies. Cytokines play a pivotal role in tumor immunotherapy by helping initiating and driving antitumor immune responses. Here, we demonstrated that, besides conventional CD4+ and CD8+ T cells, IL36 surprisingly increased the number of tumor-infiltrating regulatory T (Treg) cells in vivo and enhanced proliferation of Tregs in vitro. Administration of CTLA-4 monoclonal antibodies (mAbs) strongly enhanced IL36-stimulated antitumor activities through depletion of Tregs. In addition, a cancer gene therapy using the IL36-loaded nanoparticles in combination with CTLA-4 mAbs additively reduced lung metastasis of breast tumor cells. We further showed that the combined therapy of CTLA-4 mAbs and IL36 led to an increase in proliferation and IFN-γ production by CD4+ and CD8+ T cells when compared to single therapy with CTLA-4 mAbs or IL36. Collectively, our findings demonstrated a new combination therapy that could improve the clinical response to ICB immunotherapy for cancer.
Collapse
Affiliation(s)
- Qiuxia Qu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Zhiwei Zhai
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jieni Xu
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Song Li
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Cheng Chen
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| |
Collapse
|
22
|
Mitsi E, Carniel B, Reiné J, Rylance J, Zaidi S, Soares-Schanoski A, Connor V, Collins AM, Schlitzer A, Nikolaou E, Solórzano C, Pojar S, Hill H, Hyder-Wright AD, Jambo KC, Oggioni MR, De Ste Croix M, Gordon SB, Jochems SP, Ferreira DM. Nasal Pneumococcal Density Is Associated with Microaspiration and Heightened Human Alveolar Macrophage Responsiveness to Bacterial Pathogens. Am J Respir Crit Care Med 2020; 201:335-347. [PMID: 31626559 PMCID: PMC6999099 DOI: 10.1164/rccm.201903-0607oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Rationale: Pneumococcal pneumonia remains a global health problem. Colonization of the nasopharynx with Streptococcus pneumoniae (Spn), although a prerequisite of infection, is the main source of exposure and immunological boosting in children and adults. However, our knowledge of how nasal colonization impacts on the lung cells, especially on the predominant alveolar macrophage (AM) population, is limited.Objectives: Using a controlled human infection model to achieve nasal colonization with 6B serotype, we investigated the effect of Spn colonization on lung cells.Methods: We collected BAL from healthy pneumococcal-challenged participants aged 18-49 years. Confocal microscopy and molecular and classical microbiology were used to investigate microaspiration and pneumococcal presence in the lower airways. AM opsonophagocytic capacity was assessed by functional assays in vitro, whereas flow cytometry and transcriptomic analysis were used to assess further changes on the lung cellular populations.Measurements and Main Results: AMs from Spn-colonized individuals exhibited increased opsonophagocytosis to pneumococcus (11.4% median increase) for approximately 3 months after experimental pneumococcal colonization. AMs also had increased responses against other bacterial pathogens. Pneumococcal DNA detected in the BAL samples of Spn-colonized individuals were positively correlated with nasal pneumococcal density (r = 0.71; P = 0.029). Similarly, AM-heightened opsonophagocytic capacity was correlated with nasopharyngeal pneumococcal density (r = 0.61, P = 0.025).Conclusions: Our findings demonstrate that nasal colonization with pneumococcus and microaspiration prime AMs, leading to brisker responsiveness to both pneumococcus and unrelated bacterial pathogens. The relative abundance of AMs in the alveolar spaces, alongside their potential for nonspecific protection, render them an attractive target for novel vaccines.
Collapse
Affiliation(s)
- Elena Mitsi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Beatriz Carniel
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Jesús Reiné
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Jamie Rylance
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Seher Zaidi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Victoria Connor
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Andrea M. Collins
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Andreas Schlitzer
- The Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Elissavet Nikolaou
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Carla Solórzano
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Sherin Pojar
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Helen Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Angela D. Hyder-Wright
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Kondwani C. Jambo
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Malawi Liverpool Wellcome Trust Clinical Research Programme, College of Medicine, Chichiri, Blantyre, Malawi
| | - Marco R. Oggioni
- Department of Genetics, University of Leicester, Leicester, United Kingdom; and
| | - Megan De Ste Croix
- Department of Genetics, University of Leicester, Leicester, United Kingdom; and
| | - Stephen B. Gordon
- Malawi Liverpool Wellcome Trust Clinical Research Programme, College of Medicine, Chichiri, Blantyre, Malawi
| | - Simon P. Jochems
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Department of Parasitology, Leiden University Medical Center, Leiden, the Netherlands
| | - Daniela M. Ferreira
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
23
|
Chelvanambi M, Weinstein AM, Storkus WJ. IL-36 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1240:95-110. [PMID: 32060891 DOI: 10.1007/978-3-030-38315-2_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The ability of the immune system to prevent or control the growth of tumor cells is critically dependent on inflammatory processes that lead to the activation, expansion, and recruitment of antitumor effector cells into the tumor microenvironment (TME). These processes are orchestrated by soluble cytokines produced in tissues that alarm local immune surveillance cells (such as dendritic cells, DCs) to mobilize protective antitumor immune populations (B cells, T cells). The interleukin (IL)-36 family of pro-inflammatory cytokines plays an important role in multiple disease processes, ranging from an instigator of autoimmune psoriasis to an initiator of therapeutic immune responses against tumor cells. This chapter will focus on the biologic role of immunomodulatory IL-36 family cytokines in the cancer setting and their potential utility in the design of effective interventional therapies. (127 words).
Collapse
Affiliation(s)
- Manoj Chelvanambi
- Departments of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Aliyah M Weinstein
- Departments of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Walter J Storkus
- Departments of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Departments of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
24
|
Queen D, Ediriweera C, Liu L. Function and Regulation of IL-36 Signaling in Inflammatory Diseases and Cancer Development. Front Cell Dev Biol 2019; 7:317. [PMID: 31867327 PMCID: PMC6904269 DOI: 10.3389/fcell.2019.00317] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/20/2019] [Indexed: 12/31/2022] Open
Abstract
The IL-36 subfamily of cytokines belongs to the IL-1 superfamily and consists of three pro-inflammatory agonists IL-36α, IL-36β, IL-36γ, and an IL-36 receptor (IL-36R) antagonist, IL-36Ra. These IL-36 cytokines function through a common receptor to modulate innate and adaptive immune responses. IL-36 cytokines are expressed as inactive precursors and require proteolytic processing to become fully active. Upon binding to IL-36R, IL-36 agonists augment the expression and production of inflammatory cytokines via activating signaling pathways. IL-36 is mainly expressed in epidermal, bronchial, and intestinal epithelial cells that form the barrier structures of the body and regulates the balance between pro-inflammatory and anti-inflammatory cytokine production at these tissue sites. Dysregulation of IL-36 signaling is a major etiological factor in the development of autoimmune and inflammatory diseases. Besides its critical role in inflammatory skin diseases such as psoriasis, emerging evidence suggests that aberrant IL-36 activities also promote inflammatory diseases in the lung, kidneys, and intestines, underscoring the potential of IL-36 as a therapeutic target for common inflammatory diseases. The role of IL-36 signaling in cancer development is also under investigation, with limited studies suggesting a potential anti-tumor effect. In this comprehensive review, we summarize current knowledge regarding the expression, activation, regulatory mechanisms, and biological functions of IL-36 signaling in immunity, inflammatory diseases, and cancer development.
Collapse
Affiliation(s)
- Dawn Queen
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | | | - Liang Liu
- The Hormel Institute, University of Minnesota, Austin, MN, United States
| |
Collapse
|
25
|
Madonna S, Girolomoni G, Dinarello CA, Albanesi C. The Significance of IL-36 Hyperactivation and IL-36R Targeting in Psoriasis. Int J Mol Sci 2019; 20:E3318. [PMID: 31284527 PMCID: PMC6650959 DOI: 10.3390/ijms20133318] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/26/2019] [Accepted: 07/04/2019] [Indexed: 12/14/2022] Open
Abstract
Psoriasis is an immune-mediated inflammatory skin disease that involves mainly T helper (Th)17, Th1 and Th22 lymphocytes, which cause hyper-proliferation of the epidermis with aberrant differentiation of keratinocytes, and local production of chemokines and cytokines. These fuel a self-amplifying loop where these products act on T cells to perpetuate cutaneous inflammatory processes. Among the various inflammatory mediators involved, interleukin (IL)-36 cytokines are important for the recruitment and activation of neutrophils and Th17 cells in psoriatic skin. In particular, IL-36s induce chemokines and cytokines interfere with differentiation/cornification programs in the epidermis, as well as promote pathological angiogenesis and endothelial cell activation. IL-36 cytokines belong to the IL-1 family, and comprise IL-36α, IL-36β, and IL-36γ agonists as well as IL-36 receptor antagonist and IL-38 antagonists. IL-36 cytokines are up-regulated in psoriatic epidermis, and their expression is strongly induced by TNF-α and IL-17. Contrarily, IL-38 antagonist is downregulated, and its impaired expression may be relevant to the dysregulated inflammatory processes induced by IL-36. Here, we discuss on the pathogenic mechanisms leading to the altered balance of IL-36 agonists/antagonists and the significance of this dysregulation in psoriasis. Collection of the information will provide a theoretical basis for the development of novel therapeutic strategies based on IL-36 agonist/antagonist manipulation in psoriasis.
Collapse
Affiliation(s)
- Stefania Madonna
- Laboratory of Experimental Immunology, IDI-IRCCS, via Monti di Creta, 104, 00167 Rome, Italy.
| | - Giampiero Girolomoni
- Section of Dermatology, Department of Medicine, University of Verona, P.zza Stefani, 1, 37126 Verona, Italy
| | - Charles A Dinarello
- Department of Medicine, Radboud University Medical Center, 6525 HP Nijmegen, The Netherlands
- Department of Medicine, School of Medicine, University of Colorado, Denver 80045, Anschutz Campus, Aurora, CO, USA
| | - Cristina Albanesi
- Laboratory of Experimental Immunology, IDI-IRCCS, via Monti di Creta, 104, 00167 Rome, Italy
| |
Collapse
|
26
|
Fahey E, Doyle SL. IL-1 Family Cytokine Regulation of Vascular Permeability and Angiogenesis. Front Immunol 2019; 10:1426. [PMID: 31293586 PMCID: PMC6603210 DOI: 10.3389/fimmu.2019.01426] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/06/2019] [Indexed: 12/21/2022] Open
Abstract
The IL-1 family of cytokines are well-known for their primary role in initiating inflammatory responses both in response to and acting as danger signals. It has long been established that IL-1 is capable of simultaneously regulating inflammation and angiogenesis, indeed one of IL-1's earliest names was haemopoeitn-1 due to its pro-angiogenic effects. Other IL-1 family cytokines are also known to have roles in mediating angiogenesis, either directly or indirectly via induction of proangiogenic factors such as VEGF. Of note, some of these family members appear to have directly opposing effects in different tissues and pathologies. Here we will review what is known about how the various IL-1 family members regulate vascular permeability and angiogenic function in a range of different tissues, and describe some of the mechanisms employed to achieve these effects.
Collapse
Affiliation(s)
- Erin Fahey
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sarah L Doyle
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.,Our Lady's Children's Hospital Crumlin, National Children's Research Centre, Dublin, Ireland
| |
Collapse
|
27
|
Buhl AL, Wenzel J. Interleukin-36 in Infectious and Inflammatory Skin Diseases. Front Immunol 2019; 10:1162. [PMID: 31191535 PMCID: PMC6545975 DOI: 10.3389/fimmu.2019.01162] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 05/08/2019] [Indexed: 12/20/2022] Open
Abstract
Interleukin-36 (IL-36) comprises to a cytokine family consisting of four isoforms IL-36α, IL-36β, IL-36γ, and IL-36 receptor antagonist (IL-36 Ra). These IL-36 cytokines, in turn, belong to the IL-1 superfamily. The IL-36 receptor (IL-1R6) is functional as a heterodimer formed of IL-1R6 and IL-1 receptor accessory protein (IL-1RAcP). IL-36α, IL-36β, and IL-36γ are regarded as pro-inflammatory ligands and IL-36 Ra as well as IL-38 as anti-inflammatory ligands of IL-1R6. IL-36 cytokines are mainly expressed on the barrier sites of the body e.g., bronchial, intestinal, and dermal epithelium. One of their most important biological functions is the bridging of innate and adaptive immune responses. A disturbed balance between pro-inflammatory and anti-inflammatory branches easily leads to inflammation of the corresponding tissue. The most prominent example for an altered IL-36 expression is the spectrum of psoriasis. In addition to inflammatory dermatoses, IL-36 also seems to play a role in infectious dermatoses. Microbial triggers, especially Staphylococcus aureus infection, increase the production of pro-inflammatory IL-36 cytokines and initiate/promote the inflammation of skin lesions. Due to the discovery of IL-36 as an important immune mediator, it has already been possible to develop important diagnostic tools for dermatitis. Not only in the field of inflammatory skin diseases, but also in pulmonary and intestinal inflammation, there is evidence that IL-36 cytokines might have diagnostic and/or therapeutic relevance.
Collapse
Affiliation(s)
- Anna-Lena Buhl
- Department of Dermatology and Allergy, University Hospital of Bonn, Bonn, Germany
| | - Joerg Wenzel
- Department of Dermatology and Allergy, University Hospital of Bonn, Bonn, Germany
| |
Collapse
|
28
|
IL-36 Cytokines: Regulators of Inflammatory Responses and Their Emerging Role in Immunology of Reproduction. Int J Mol Sci 2019; 20:ijms20071649. [PMID: 30987081 PMCID: PMC6479377 DOI: 10.3390/ijms20071649] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 03/28/2019] [Accepted: 03/28/2019] [Indexed: 01/01/2023] Open
Abstract
The IL-36 subfamily of cytokines has been recently described as part of the IL-1 superfamily. It comprises three pro-inflammatory agonists (IL-36α, IL-36β, and IL-36γ), their receptor (IL-36R), and one antagonist (IL-36Ra). Although expressed in a variety of cells, the biological relevance of IL-36 cytokines is most evident in the communication between epithelial cells, dendritic cells, and neutrophils, which constitute the common triad responsible for the initiation, maintenance, and expansion of inflammation. The immunological role of IL-36 cytokines was initially described in studies of psoriasis, but novel evidence demonstrates their involvement in further immune and inflammatory processes in physiological and pathological situations. Preliminary studies have reported a dynamic expression of IL-36 cytokines in the female reproductive tract throughout the menstrual cycle, as well as their association with the production of immune mediators and cellular recruitment in the vaginal microenvironment contributing to host defense. In pregnancy, alteration of the placental IL-36 axis has been reported upon infection and pre-eclampsia suggesting its pivotal role in the regulation of maternal immune responses. In this review, we summarize current knowledge regarding the regulatory mechanisms and biological actions of IL-36 cytokines, their participation in different inflammatory conditions, and the emerging data on their potential role in normal and complicated pregnancies.
Collapse
|
29
|
IL-36, IL-37, and IL-38 Cytokines in Skin and Joint Inflammation: A Comprehensive Review of Their Therapeutic Potential. Int J Mol Sci 2019; 20:ijms20061257. [PMID: 30871134 PMCID: PMC6470667 DOI: 10.3390/ijms20061257] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 02/06/2023] Open
Abstract
The interleukin (IL)-1 family of cytokines is composed of 11 members, including the most recently discovered IL-36α, β, γ, IL-37, and IL-38. Similar to IL-1, IL-36 cytokines are initiators and amplifiers of inflammation, whereas both IL-37 and IL-38 display anti-inflammatory activities. A few studies have outlined the role played by these cytokines in several inflammatory diseases. For instance, IL-36 agonists seem to be relevant for the pathogenesis of skin psoriasis whereas, despite being expressed within the synovial tissue, their silencing or overexpression do not critically influence the course of arthritis in mice. In this review, we will focus on the state of the art of the molecular features and biological roles of IL-36, IL-37, and IL-38 in representative skin- and joint-related inflammatory diseases, namely psoriasis, rheumatoid arthritis, and psoriatic arthritis. We will then offer an overview of the therapeutic potential of targeting the IL-36 axis in these diseases, either by blocking the proinflammatory agonists or enhancing the physiologic inhibitory feedback on the inflammation mediated by the antagonists IL-37 and IL-38.
Collapse
|
30
|
Liu S, Wu F, Wu Z, Li Y, Zhang S, Yu N. IL-17A synergistically enhances TLR3-mediated IL-36γ production by keratinocytes: A potential role in injury-amplified psoriatic inflammation. Exp Dermatol 2019; 28:233-239. [PMID: 30614571 DOI: 10.1111/exd.13871] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 12/18/2018] [Accepted: 01/04/2019] [Indexed: 12/11/2022]
Abstract
Skin injury can trigger formation of new lesions in psoriasis (Koebner phenomenon). The mechanisms through which injury exacerbates psoriasis are unclear. During wound repair, epidermal keratinocytes are activated and produce abundant IL-36γ, further promoting the skin inflammation. IL-17A is the cornerstone cytokine in the pathogenesis of psoriasis. We sought to investigate the effects of IL-17A on injury-induced keratinocyte activation and IL-36γ production. Here, we demonstrated that dsRNA released from necrotic keratinocytes induced the expression of IL-36γ. Silencing of TLR3 by siRNA decreased the IL-36γ induction by necrotic keratinocyte supernatant. Co-stimulation with dsRNA and IL-17A synergistically increased the expression of IL-36γ and other proinflammatory mediators (CCL20, CXCL8, DEFB4 and LCN2) in keratinocytes. The synergistic effects were not dependent on TLR3 upregulation, TNF receptor signalling and mRNA stabilization. Co-stimulation with dsRNA and IL-17A resulted in an accumulation of IκBζ. The synergistic upregulation of IL-36γ and proinflammatory mediators were inhibited by IκBζ siRNA. Co-stimulation with IL-17A and poly(I:C) markedly activated the p38 MAPK and NF-κB pathway, compared with poly(I:C). Blockade of p38 MAPK and NF-κB suppressed dsRNA/IL-17A-mediated IκBζ and IL-36γ induction. These findings demonstrated that IL-17A synergistically enhanced the dsRNA-mediated IL-36γ production through a p38 MAPK-, NF-κB-, and IκBζ-dependent mechanism.
Collapse
Affiliation(s)
- Shuangchun Liu
- Department of Blood Transfusion, Taizhou Municipal Hospital, Taizhou, China
| | - Fei Wu
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Zongzhou Wu
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Ying Li
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Shujie Zhang
- Shanghai Key Laboratory of Visual Impairment and Restoration, Eye Institute, Eye and ENT Hospital of Fudan University, Shanghai, China
| | - Ning Yu
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| |
Collapse
|
31
|
Overlapping Roles for Interleukin-36 Cytokines in Protective Host Defense against Murine Legionella pneumophila Pneumonia. Infect Immun 2018; 87:IAI.00583-18. [PMID: 30323031 PMCID: PMC6300640 DOI: 10.1128/iai.00583-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/12/2018] [Indexed: 01/19/2023] Open
Abstract
Legionella pneumophila causes life-threatening pneumonia culminating in acute lung injury. Innate and adaptive cytokines play an important role in host defense against L. pneumophila infection. Interleukin-36 (IL-36) cytokines are recently described members of the larger IL-1 cytokine family known to exert potent inflammatory effects. In this study, we elucidated the role for IL-36 cytokines in experimental pneumonia caused by L. pneumophila Intratracheal (i.t.) administration of L. pneumophila induced the upregulation of both IL-36α and IL-36γ mRNA and protein production in the lung. Compared to the findings for L. pneumophila-infected wild-type (WT) mice, the i.t. administration of L. pneumophila to IL-36 receptor-deficient (IL-36R-/-) mice resulted in increased mortality, a delay in lung bacterial clearance, increased L. pneumophila dissemination to extrapulmonary organs, and impaired glucose homeostasis. Impaired lung bacterial clearance in IL-36R-/- mice was associated with a significantly reduced accumulation of inflammatory cells and the decreased production of proinflammatory cytokines and chemokines. Ex vivo, reduced expression of costimulatory molecules and impaired M1 polarization were observed in alveolar macrophages isolated from infected IL-36R-/- mice compared to macrophages from WT mice. While L. pneumophila-induced mortality in IL-36α- or IL-36γ-deficient mice was not different from that in WT animals, antibody-mediated neutralization of IL-36γ in IL-36α-/- mice resulted in mortality similar to that observed in IL-36R-/- mice, indicating redundant and overlapping roles for these cytokines in experimental murine L. pneumophila pneumonia.
Collapse
|
32
|
Leal-Gutiérrez JD, Rezende FM, Elzo MA, Johnson D, Peñagaricano F, Mateescu RG. Structural Equation Modeling and Whole-Genome Scans Uncover Chromosome Regions and Enriched Pathways for Carcass and Meat Quality in Beef. Front Genet 2018; 9:532. [PMID: 30555508 PMCID: PMC6282042 DOI: 10.3389/fgene.2018.00532] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/22/2018] [Indexed: 12/11/2022] Open
Abstract
Structural equation models involving latent variables are useful tools for formulating hypothesized models defined by theoretical variables and causal links between these variables. The objectives of this study were: (1) to identify latent variables underlying carcass and meat quality traits and (2) to perform whole-genome scans for these latent variables in order to identify genomic regions and individual genes with both direct and indirect effects. A total of 726 steers from an Angus-Brahman multibreed population with records for 22 phenotypes were used. A total of 480 animals were genotyped with the GGP Bovine F-250. The single-step genomic best linear unbiased prediction method was used to estimate the amount of genetic variance explained for each latent variable by chromosome regions of 20 adjacent SNP-windows across the genome. Three types of genetic effects were considered: (1) direct effects on a single latent phenotype; (2) direct effects on two latent phenotypes simultaneously; and (3) indirect effects. The final structural model included carcass quality as an independent latent variable and meat quality as a dependent latent variable. Carcass quality was defined by quality grade, fat over the ribeye and marbling, while the meat quality was described by juiciness, tenderness and connective tissue, all of them measured through a taste panel. From 571 associated genomic regions (643 genes), each one explaining at least 0.05% of the additive variance, 159 regions (179 genes) were associated with carcass quality, 106 regions (114 genes) were associated with both carcass and meat quality, 242 regions (266 genes) were associated with meat quality, and 64 regions (84 genes) were associated with carcass quality, having an indirect effect on meat quality. Three biological mechanisms emerged from these findings: postmortem proteolysis of structural proteins and cellular compartmentalization, cellular proliferation and differentiation of adipocytes, and fat deposition.
Collapse
Affiliation(s)
| | - Fernanda M. Rezende
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
- Faculdade de Medicina Veterinária, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Mauricio A. Elzo
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Dwain Johnson
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Francisco Peñagaricano
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
- University of Florida Genetics Institute, University of Florida, Gainesville, FL, United States
| | - Raluca G. Mateescu
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| |
Collapse
|
33
|
Weinstein AM, Giraldo NA, Petitprez F, Julie C, Lacroix L, Peschaud F, Emile JF, Marisa L, Fridman WH, Storkus WJ, Sautès-Fridman C. Association of IL-36γ with tertiary lymphoid structures and inflammatory immune infiltrates in human colorectal cancer. Cancer Immunol Immunother 2018; 68:109-120. [PMID: 30315348 DOI: 10.1007/s00262-018-2259-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 10/04/2018] [Indexed: 02/07/2023]
Abstract
IL-1 family cytokines play a dual role in the gut, with different family members contributing either protective or pathogenic effects. IL-36γ is an IL-1 family cytokine involved in polarizing type-1 immune responses. However, its function in the gut, including in colorectal cancer pathogenesis, is not well appreciated. In a murine model of colon carcinoma, IL-36γ controls tertiary lymphoid structure formation and promotes a type-1 immune response concurrently with a decrease in expression of immune checkpoint molecules in the tumor microenvironment. Here, we demonstrate that IL-36γ plays a similar role in driving a pro-inflammatory phenotype in human colorectal cancer. We analyzed a cohort of 33 primary colorectal carcinoma tumors using imaging, flow cytometry, and transcriptomics to determine the pattern and role of IL-36γ expression in this disease. In the colorectal tumor microenvironment, we observed IL-36γ to be predominantly expressed by M1 macrophages and cells of the vasculature, including smooth muscle cells and high endothelial venules. This pattern of IL-36γ expression is associated with a CD4+ central memory T cell infiltrate and an increased density of B cells in tertiary lymphoid structures, as well as with markers of fibrosis. Conversely, expression of the antagonist to IL-36 signaling, IL-1F5, was associated with intratumoral expression of checkpoint molecules, including PD-1, PD-L1, and CTLA4, which can suppress the immune response. These data support a role for IL-36γ in the physiologic immune response to colorectal cancer by sustaining inflammation within the tumor microenvironment.
Collapse
Affiliation(s)
- Aliyah M Weinstein
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, Immune Control and Escape", 75006, Paris, France.
- University Paris Descartes, Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France.
- Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France.
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Nicolas A Giraldo
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, Immune Control and Escape", 75006, Paris, France
- University Paris Descartes, Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
- Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
- Pathology Department, Johns Hopkins Hospital, Baltimore, MD, 21287, USA
| | - Florent Petitprez
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, Immune Control and Escape", 75006, Paris, France
- University Paris Descartes, Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
- Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
- Programme Cartes d'Identités des Tumeurs, Ligue Nationale contre le Cancer, 75013, Paris, France
| | - Catherine Julie
- Laboratoire d'anatomie pathologique, Hopital Ambroise Paré, AP-HP, Boulogne, France
- EA4340, Université de Versailles SQY, Université Paris Saclay, Boulogne, France
| | - Laetitia Lacroix
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, Immune Control and Escape", 75006, Paris, France
- University Paris Descartes, Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
- Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
| | - Frédérique Peschaud
- Laboratoire d'anatomie pathologique, Hopital Ambroise Paré, AP-HP, Boulogne, France
- EA4340, Université de Versailles SQY, Université Paris Saclay, Boulogne, France
| | - Jean-François Emile
- Laboratoire d'anatomie pathologique, Hopital Ambroise Paré, AP-HP, Boulogne, France
- EA4340, Université de Versailles SQY, Université Paris Saclay, Boulogne, France
| | - Laetitia Marisa
- Programme Cartes d'Identités des Tumeurs, Ligue Nationale contre le Cancer, 75013, Paris, France
| | - Wolf H Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, Immune Control and Escape", 75006, Paris, France
- University Paris Descartes, Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
- Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
| | - Walter J Storkus
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Pathology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Bioengineering, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Catherine Sautès-Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, Immune Control and Escape", 75006, Paris, France
- University Paris Descartes, Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
- Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
| |
Collapse
|
34
|
Bassoy EY, Towne JE, Gabay C. Regulation and function of interleukin-36 cytokines. Immunol Rev 2018; 281:169-178. [PMID: 29247994 DOI: 10.1111/imr.12610] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The interleukin (IL)-36 cytokines include 3 agonists, IL-36α, IL-36β, and IL-36γ that bind to a common receptor composed of IL-36R and IL-1RAcP to stimulate inflammatory responses. IL-36Ra is a natural antagonist that binds to IL-36R, but does not recruit the co-receptor IL-1RAcP and does not stimulate any intracellular responses. The IL-36 cytokines are expressed predominantly by epithelial cells and act on a number of cells including immune cells, epithelial cells, and fibroblasts. Processing of the N-terminus is required for full agonist or antagonist activity for all IL-36 members. The role of IL-36 has been extensively demonstrated in the skin where it can act on keratinocytes and immune cells to induce a robust inflammatory response that has been implicated in psoriatic disorders. Emerging data also suggest a role for this cytokine family in pulmonary and intestinal physiology and pathology.
Collapse
Affiliation(s)
- Esen Yonca Bassoy
- Division of Rheumatology, Department of Internal Medicine Specialties & Department of Pathology-Immunology, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland
| | - Jennifer E Towne
- Immunology Discovery, Janssen Research and Development, San Diego, CA, USA
| | - Cem Gabay
- Division of Rheumatology, Department of Internal Medicine Specialties & Department of Pathology-Immunology, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland
| |
Collapse
|
35
|
Robinson KM, Ramanan K, Clay ME, McHugh KJ, Pilewski MJ, Nickolich KL, Corey C, Shiva S, Wang J, Muzumdar R, Alcorn JF. The inflammasome potentiates influenza/Staphylococcus aureus superinfection in mice. JCI Insight 2018; 3:97470. [PMID: 29618653 DOI: 10.1172/jci.insight.97470] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 02/28/2018] [Indexed: 01/24/2023] Open
Abstract
Secondary bacterial respiratory infections are commonly associated with both acute and chronic lung injury. Influenza complicated by bacterial pneumonia is an effective model to study host defense during pulmonary superinfection due to its clinical relevance. Multiprotein inflammasomes are responsible for IL-1β production in response to infection and drive tissue inflammation. In this study, we examined the role of the inflammasome during viral/bacterial superinfection. We demonstrate that ASC-/- mice are protected from bacterial superinfection and produce sufficient quantities of IL-1β through an apoptosis-associated speck-like protein containing CARD (ASC) inflammasome-independent mechanism. Despite the production of IL-1β by ASC-/- mice in response to bacterial superinfection, these mice display decreased lung inflammation. A neutrophil elastase inhibitor blocked ASC inflammasome-independent production of IL-1β and the IL-1 receptor antagonist, anakinra, confirmed that IL-1 remains crucial to the clearance of bacteria during superinfection. Delayed inhibition of NLRP3 during influenza infection by MCC950 decreases bacterial burden during superinfection and leads to decreased inflammatory cytokine production. Collectively, our results demonstrate that ASC augments the clearance of bacteria, but can also contribute to inflammation and mortality. ASC should be considered as a therapeutic target to decrease morbidity and mortality during bacterial superinfection.
Collapse
Affiliation(s)
- Keven M Robinson
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Krishnaveni Ramanan
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Michelle E Clay
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Kevin J McHugh
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | - Matthew J Pilewski
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kara L Nickolich
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Catherine Corey
- Department of Pharmacology and Chemical Biology, and.,Vascular Medical Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, and.,Vascular Medical Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jieru Wang
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | - Radhika Muzumdar
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | - John F Alcorn
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
36
|
Nishikawa H, Taniguchi Y, Matsumoto T, Arima N, Masaki M, Shimamura Y, Inoue K, Horino T, Fujimoto S, Ohko K, Komatsu T, Udaka K, Sano S, Terada Y. Knockout of the interleukin-36 receptor protects against renal ischemia-reperfusion injury by reduction of proinflammatory cytokines. Kidney Int 2018; 93:599-614. [DOI: 10.1016/j.kint.2017.09.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 08/23/2017] [Accepted: 09/14/2017] [Indexed: 10/18/2022]
|
37
|
IL-36γ is a crucial proximal component of protective type-1-mediated lung mucosal immunity in Gram-positive and -negative bacterial pneumonia. Mucosal Immunol 2017; 10:1320-1334. [PMID: 28176791 PMCID: PMC5548659 DOI: 10.1038/mi.2016.130] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 12/01/2016] [Indexed: 02/04/2023]
Abstract
Interleukin-36γ (IL-36γ) is a member of novel IL-1-like proinflammatory cytokine family that are highly expressed in epithelial tissues and several myeloid-derived cell types. Little is known about the role of the IL-36 family in mucosal immunity, including lung anti-bacterial responses. We used murine models of IL-36γ deficiency to assess the contribution of IL-36γ in the lung during experimental pneumonia. Induction of IL-36γ was observed in the lung in response to Streptococcus pneumoniae (Sp) infection, and mature IL-36γ protein was secreted primarily in microparticles. IL-36γ-deficient mice challenged with Sp demonstrated increased mortality, decreased lung bacterial clearance and increased bacterial dissemination, in association with reduced local expression of type-1 cytokines, and impaired lung macrophage M1 polarization. IL-36γ directly stimulated type-1 cytokine induction from dendritic cells in vitro in a MyD88-dependent manner. Similar protective effects of IL-36γ were observed in a Gram-negative pneumonia model (Klebsiella pneumoniae). Intrapulmonary delivery of IL-36γ-containing microparticles reconstituted immunity in IL-36γ-/- mice. Enhanced expression of IL-36γ was also observed in plasma and bronchoalveolar lavage fluid of patients with acute respiratory distress syndrome because of pneumonia. These studies indicate that IL-36γ assumes a vital proximal role in the lung innate mucosal immunity during bacterial pneumonia by driving protective type-1 responses and classical macrophage activation.
Collapse
|
38
|
Jensen LE. Interleukin-36 cytokines may overcome microbial immune evasion strategies that inhibit interleukin-1 family signaling. Sci Signal 2017; 10:10/492/eaan3589. [DOI: 10.1126/scisignal.aan3589] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
39
|
Weinstein AM, Chen L, Brzana EA, Patil PR, Taylor JL, Fabian KL, Wallace CT, Jones SD, Watkins SC, Lu B, Stroncek DF, Denning TL, Fu YX, Cohen PA, Storkus WJ. Tbet and IL-36γ cooperate in therapeutic DC-mediated promotion of ectopic lymphoid organogenesis in the tumor microenvironment. Oncoimmunology 2017; 6:e1322238. [PMID: 28680760 DOI: 10.1080/2162402x.2017.1322238] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 12/31/2022] Open
Abstract
We have previously reported that direct injection of dendritic cells (DC) engineered to express the Type-1 transactivator Tbet (i.e., DC.Tbet) into murine tumors results in antitumor efficacy in association with the development of structures resembling tertiary lymphoid organs (TLO) in the tumor microenvironment (TME). These TLO contained robust infiltrates of B cells, DC, NK cells, and T cells in proximity to PNAd+ blood vessels; however, they were considered incomplete, since the recruited B cells failed to organize into classic germinal center-like structures. We now report that antitumor efficacy and TLO-inducing capacity of DC.Tbet-based i.t. therapy is operational in peripheral lymph node-deficient LTA-/- mice, and that it is highly dependent upon a direct Tbet target gene product, IL-36γ/IL-1F9. Intratumoral DC.Tbet fails to provide protection to tumor-bearing IL-36R-/- hosts, or to tumor-bearing wild-type recipient mice co-administered rmIL-1F5/IL-36RN, a natural IL-36R antagonist. Remarkably, the injection of tumors with DC engineered to secrete a bioactive form of mIL-36γ (DC.IL36γ) also initiated therapeutic TLO and slowed tumor progression in vivo. Furthermore, DC.IL36γ cells strongly upregulated their expression of Tbet, suggesting that Tbet and IL-36γ cooperate to reinforce each other's expression in DC, rendering them competent to promote TLO formation in an "immunologically normalized," therapeutic TME.
Collapse
Affiliation(s)
- Aliyah M Weinstein
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
| | - Lu Chen
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
| | | | | | | | - Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
| | - Callen T Wallace
- Department of Cell Biology and Physiology, UPSOM, Pittsburgh, PA, USA
| | | | - Simon C Watkins
- Department of Cell Biology and Physiology, UPSOM, Pittsburgh, PA, USA
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
| | - David F Stroncek
- Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, PA, USA
| | - Timothy L Denning
- Center for Inflammation, Immunity & Infection at Georgia State University, Atlanta, GA, USA
| | - Yang-Xin Fu
- Departments of Pathology and Immunology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Peter A Cohen
- Department of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA.,Department of Dermatology, UPSOM, Pittsburgh, PA, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Bioengineering, UPSOM, Pittsburgh, PA, USA.,University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
40
|
Conway C, Graham JL, Chengot P, Daly C, Chalkley R, Ross L, Droop A, Rabbitts P, Stead LF. Elucidating drivers of oral epithelial dysplasia formation and malignant transformation to cancer using RNAseq. Oncotarget 2016; 6:40186-201. [PMID: 26515596 PMCID: PMC4741888 DOI: 10.18632/oncotarget.5529] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 10/09/2015] [Indexed: 02/07/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a prevalent cancer with poor prognosis. Most OSCC progresses via a non-malignant stage called dysplasia. Effective treatment of dysplasia prior to potential malignant transformation is an unmet clinical need. To identify markers of early disease, we performed RNA sequencing of 19 matched HPV negative patient trios: normal oral mucosa, dysplasia and associated OSCC. We performed differential gene expression, principal component and correlated gene network analysis using these data. We found differences in the immune cell signatures present at different disease stages and were able to distinguish early events in pathogenesis, such as upregulation of many HOX genes, from later events, such as down-regulation of adherens junctions. We herein highlight novel coding and non-coding candidates for involvement in oral dysplasia development and malignant transformation, and speculate on how our findings may guide further translational research into the treatment of oral dysplasia.
Collapse
Affiliation(s)
- Caroline Conway
- Precancer Genomics, Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, UK.,Stratified Medicine (Oncology), School of Biomedical Sciences, University of Ulster, Coleraine, Co. Londonderry, BT52 1SA, UK
| | - Jennifer L Graham
- Precancer Genomics, Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, UK
| | - Preetha Chengot
- Precancer Genomics, Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, UK
| | - Catherine Daly
- Precancer Genomics, Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, UK
| | - Rebecca Chalkley
- Precancer Genomics, Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, UK
| | - Lisa Ross
- Precancer Genomics, Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, UK
| | - Alastair Droop
- Precancer Genomics, Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, UK
| | - Pamela Rabbitts
- Precancer Genomics, Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, UK
| | - Lucy F Stead
- Precancer Genomics, Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, UK
| |
Collapse
|
41
|
Interleukin-36 potently stimulates human M2 macrophages, Langerhans cells and keratinocytes to produce pro-inflammatory cytokines. Cytokine 2016; 84:88-98. [DOI: 10.1016/j.cyto.2016.05.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/10/2016] [Accepted: 05/10/2016] [Indexed: 12/11/2022]
|
42
|
Ahsan F, Moura-Alves P, Guhlich-Bornhof U, Klemm M, Kaufmann SHE, Maertzdorf J. Role of Interleukin 36γ in Host Defense Against Tuberculosis. J Infect Dis 2016; 214:464-74. [DOI: 10.1093/infdis/jiw152] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/07/2016] [Indexed: 12/11/2022] Open
|
43
|
Boutet MA, Bart G, Penhoat M, Amiaud J, Brulin B, Charrier C, Morel F, Lecron JC, Rolli-Derkinderen M, Bourreille A, Vigne S, Gabay C, Palmer G, Le Goff B, Blanchard F. Distinct expression of interleukin (IL)-36α, β and γ, their antagonist IL-36Ra and IL-38 in psoriasis, rheumatoid arthritis and Crohn's disease. Clin Exp Immunol 2016; 184:159-73. [PMID: 26701127 DOI: 10.1111/cei.12761] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2015] [Indexed: 12/11/2022] Open
Abstract
Interleukin (IL)-36α, IL-36β and IL-36γ are expressed highly in skin and are involved in the pathogenesis of psoriasis, while the antagonists IL-36Ra or IL-38, another potential IL-36 inhibitor, limit uncontrolled inflammation. The expression and role of IL-36 cytokines in rheumatoid arthritis (RA) and Crohn's disease (CD) is currently debated. Here, we observed that during imiquimod-induced mouse skin inflammation and in human psoriasis, expression of IL-36α, γ and IL-36Ra, but not IL-36β and IL-38 mRNA, was induced and correlated with IL-1β and T helper type 17 (Th17) cytokines (IL-17A, IL-22, IL-23, CCL20). In mice with collagen-induced arthritis and in the synovium of patients with RA, IL-36α, β, γ, IL-36Ra and IL-38 were all elevated and correlated with IL-1β, CCL3, CCL4 and macrophage colony-stimulating factor (M-CSF), but not with Th17 cytokines. In the colon of mice with dextran sulphate sodium-induced colitis and in patients with CD, only IL-36α, γ and IL-38 were induced at relatively low levels and correlated with IL-1β and IL-17A. We suggest that only a minor subgroup of patients with RA (17-29%) or CD (25%) had an elevated IL-36 agonists/antagonists ratio, versus 93% of patients with psoriasis. By immunohistochemistry, IL-36 cytokines were produced by various cell types in skin, synovium and colonic mucosa such as keratinocytes, CD68⁺ macrophages, dendritic/Langerhans cells and CD79α⁺ plasma cells. In primary cultures of monocytes or inflammatory macrophages (M1), IL-36β and IL-36Ra were produced constitutively, but IL-36α, γ and IL-38 were produced after lipopolysaccharide stimulation. These distinct expression profiles may help to explain why only subgroups of RA and CD patients have a potentially elevated IL-36 agonists/antagonists ratio.
Collapse
Affiliation(s)
- M-A Boutet
- INSERM, UMR 957, Nantes, France.,Laboratoire De Physiopathologie De La Résorption Osseuse, Faculté De Médecine, Université De Nantes, Nantes Atlantique Universités
| | - G Bart
- INSERM, UMR 957, Nantes, France.,Laboratoire De Physiopathologie De La Résorption Osseuse, Faculté De Médecine, Université De Nantes, Nantes Atlantique Universités.,Rheumatology Unit, Nantes University Hospital, Nantes, France
| | - M Penhoat
- INSERM, UMR 957, Nantes, France.,Laboratoire De Physiopathologie De La Résorption Osseuse, Faculté De Médecine, Université De Nantes, Nantes Atlantique Universités.,Rheumatology Unit, Nantes University Hospital, Nantes, France
| | - J Amiaud
- INSERM, UMR 957, Nantes, France.,Laboratoire De Physiopathologie De La Résorption Osseuse, Faculté De Médecine, Université De Nantes, Nantes Atlantique Universités
| | - B Brulin
- INSERM, UMR 957, Nantes, France.,Laboratoire De Physiopathologie De La Résorption Osseuse, Faculté De Médecine, Université De Nantes, Nantes Atlantique Universités
| | - C Charrier
- INSERM, UMR 957, Nantes, France.,Laboratoire De Physiopathologie De La Résorption Osseuse, Faculté De Médecine, Université De Nantes, Nantes Atlantique Universités
| | - F Morel
- EA 4331, University of Poitiers, Poitiers, France
| | - J-C Lecron
- EA 4331, University of Poitiers, Poitiers, France.,Service Immunologie/Inflammation, Poitiers University Hospital, Poitiers, France
| | | | - A Bourreille
- INSERM, UMR 913, Nantes, France.,Service d'Hépato-Gastroentérologie, Nantes University Hospital, Nantes, France
| | - S Vigne
- Division of Rheumatology, Department of Internal Medicine Specialties, University Hospitals of Geneva and Department of Pathology-Immunology, University of Geneva School of Medicine, Geneva, Switzerland
| | - C Gabay
- Division of Rheumatology, Department of Internal Medicine Specialties, University Hospitals of Geneva and Department of Pathology-Immunology, University of Geneva School of Medicine, Geneva, Switzerland
| | - G Palmer
- Division of Rheumatology, Department of Internal Medicine Specialties, University Hospitals of Geneva and Department of Pathology-Immunology, University of Geneva School of Medicine, Geneva, Switzerland
| | - B Le Goff
- INSERM, UMR 957, Nantes, France.,Laboratoire De Physiopathologie De La Résorption Osseuse, Faculté De Médecine, Université De Nantes, Nantes Atlantique Universités.,Rheumatology Unit, Nantes University Hospital, Nantes, France
| | - F Blanchard
- INSERM, UMR 957, Nantes, France.,Laboratoire De Physiopathologie De La Résorption Osseuse, Faculté De Médecine, Université De Nantes, Nantes Atlantique Universités
| |
Collapse
|
44
|
Kovach MA, Singer BH, Newstead MW, Zeng X, Moore TA, White ES, Kunkel SL, Peters-Golden M, Standiford TJ. IL-36γ is secreted in microparticles and exosomes by lung macrophages in response to bacteria and bacterial components. J Leukoc Biol 2016; 100:413-21. [PMID: 26864267 DOI: 10.1189/jlb.4a0315-087r] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 01/22/2016] [Indexed: 11/24/2022] Open
Abstract
Interleukin-36 is a family of novel interleukin-1-like proinflammatory cytokines that are highly expressed in epithelial tissues and several myeloid-derived cell types. Like those of classic interleukin-1 cytokines, the secretion mechanisms of interleukin-36 are not well understood. Interleukin-36γ secretion in dermal epithelial cells requires adenosine 5'-triphosphate, which suggests a nonclassical mechanism of secretion. In this study, murine pulmonary macrophages and human alveolar macrophages were treated with recombinant pathogen-associated molecular patterns (intact bacteria: Klebsiella pneumoniae or Streptococcus pneumoniae). Cell lysates were analyzed for messenger ribonucleic acid by quantitative real-time polymerase chain reaction, and conditioned medium was analyzed for interleukin-36γ by enzyme-linked immunosorbent assay, with or without sonication. In addition, conditioned medium was ultracentrifuged at 25,000 g and 100,000 g, to isolate microparticles and exosomes, respectively, and interleukin-36γ protein was assessed in each fraction by Western blot analysis. Interleukin-36γ mRNA was induced in both murine and human lung macrophages by a variety of pathogen-associated molecular patterns, as well as heat-killed and live Klebsiella pneumoniae and Streptococcus pneumoniae, and induction occurred in a myeloid differentiation response gene 88-dependent manner. Secretion of interleukin-36γ protein was enhanced by adenosine 5'-triphosphate. Furthermore, extracellular interleukin-36γ protein detection was markedly enhanced by sonication to disrupt membrane-bound structures. Interleukin-36γ protein was detected by Western blot in microparticles and exosome fractions isolated by ultracentrifugation. Interleukin-36γ was induced and secreted from lung macrophages in response to Gram-negative and -positive bacterial stimulation. The results suggest that interleukin-36γ is secreted in a non-Golgi-dependent manner by lung macrophages in response to Gram-positive and -negative bacterial challenge.
Collapse
Affiliation(s)
- Melissa A Kovach
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine
| | - Benjamin H Singer
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine
| | - Michael W Newstead
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine
| | - Xianying Zeng
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine
| | - Thomas A Moore
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine
| | - Eric S White
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine
| | - Steven L Kunkel
- Department of Pathology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Marc Peters-Golden
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine
| | | |
Collapse
|
45
|
Huynh J, Scholz GM, Aw J, Kwa MQ, Achuthan A, Hamilton JA, Reynolds EC. IRF6 Regulates the Expression of IL-36γ by Human Oral Epithelial Cells in Response to Porphyromonas gingivalis. THE JOURNAL OF IMMUNOLOGY 2016; 196:2230-8. [PMID: 26819203 DOI: 10.4049/jimmunol.1501263] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 12/21/2015] [Indexed: 01/13/2023]
Abstract
IFN regulatory factors (IRFs) help to shape the immune response to pathogens by imparting signaling specificity to individual TLRs. We recently demonstrated that IRF6 provides specificity to TLR2 signaling in oral epithelial cells. TLR2 plays an important role in eliciting inflammation to Porphyromonas gingivalis, a keystone pathogen in periodontitis. Therefore, we investigated a role for IRF6 in mediating the inflammatory cytokine response of oral epithelial cells to P. gingivalis. IRF6 expression was strongly upregulated when human oral epithelial cells were challenged with P. gingivalis. Moreover, gene silencing and gene promoter experiments indicated that IRF6 acts downstream of IL-1R-associated kinase 1 to stimulate the expression of the IL-1 family cytokine IL-36γ in response to P. gingivalis. IRF6 and IL-1R-associated kinase 1 also regulated the stimulation of IL-36γ expression by a TLR2 agonist. IL-36γ was shown to elicit inflammatory responses by human monocyte-derived dendritic cells and macrophages, including the expression of the neutrophil chemokines IL-8 and CXCL1, as well as the Th17 chemokine CCL20. IL-36γ similarly stimulated their expression by human oral epithelial cells. Significantly, the Th17 cytokine IL-17 not only stimulated the expression of important regulators of neutrophil recruitment and survival by oral epithelial cells, but IL-17 also stimulated them to express IL-36γ. Thus, our findings suggest that IRF6 is likely to promote inflammation to P. gingivalis through its regulation of IL-36γ.
Collapse
Affiliation(s)
- Jennifer Huynh
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia; and
| | - Glen M Scholz
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia; and
| | - Jiamin Aw
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia; and
| | - Mei Qi Kwa
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia; and Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Adrian Achuthan
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - John A Hamilton
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Eric C Reynolds
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia; and
| |
Collapse
|
46
|
Interleukin (IL)-1β Is a Strong Inducer of IL-36γ Expression in Human Colonic Myofibroblasts. PLoS One 2015; 10:e0138423. [PMID: 26562662 PMCID: PMC4643060 DOI: 10.1371/journal.pone.0138423] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 08/30/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUNDS AND AIMS Interleukin (IL)-36 cytokines are members of the IL-1 cytokine family. In this study, we investigated the expression of IL-36γ in human colonic myofibroblasts to explore the molecular mechanisms underlying IL-36γ induction. MATERIALS AND METHODS IL-36 mRNA was analyzed by real-time PCR method. Secretion of IL-36γ protein was evaluated by Western blot and ELISA analyses. Molecular mechanism of IL-36γ induction was evaluated by siRNA analyses and immunofluorescence experiments. RESULTS IL-36γ mRNA expression was scarcely detected in the cells without stimulation. IL-1β induced a marked increase of IL-36γ mRNA expression. TNF-α markedly enhanced IL-1β-induced IL-36γ mRNA expression. These responses were confirmed at the protein levels. The inhibitors for ERK1/2 (PD98059 and U0216) and a p38 MAPK (SB203580) significantly reduced the IL-1β-induced IL-36γ mRNA expression. In addition, the siRNAs specific for NF-κB p65 and AP-1 (c-Jun) significantly reduced the expression of IL-1β-induced IL-36γ mRNA. CONCLUSIONS Colonic myofibroblasts are cellular source of IL-36γ in the intestine. IL-36γ expression was induced by the combination of IL-1β and TNF-α via activation of MAPKs and transcription factors, NF-κB and AP-1.
Collapse
|
47
|
Interleukin-36γ is expressed by neutrophils and can activate microglia, but has no role in experimental autoimmune encephalomyelitis. J Neuroinflammation 2015; 12:173. [PMID: 26377915 PMCID: PMC4574267 DOI: 10.1186/s12974-015-0392-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/03/2015] [Indexed: 12/25/2022] Open
Abstract
Background Experimental autoimmune encephalomyelitis (EAE) is a model of inflammatory demyelinating diseases mediated by different types of leukocytes. How these cells communicate with each other to orchestrate autoimmune attacks is not fully understood, especially in the case of neutrophils, whose importance in EAE is newly established. The present study aimed to determine the expression pattern and role of different components of the IL-36 signaling pathway (IL-36α, IL-36β, IL-36γ, IL-36R) in EAE. Methods EAE was induced by either active immunization with myelin peptide, passive transfer of myelin-reactive T cells or injection of pertussis toxin to transgenic 2D2 mice. The molecules of interest were analyzed using a combination of techniques, including quantitative real-time PCR (qRT-PCR), flow cytometry, Western blotting, in situ hybridization, and immunohistochemistry. Microglial cultures were treated with recombinant IL-36γ and analyzed using DNA microarrays. Different mouse strains were subjected to clinical evaluation and flow cytometric analysis in order to compare their susceptibility to EAE. Results Our observations indicate that both IL-36γ and IL-36R are strongly upregulated in nervous and hematopoietic tissues in different forms of EAE. IL-36γ is specifically expressed by neutrophils, while IL-36R is expressed by different immune cells, including microglia and other myeloid cells. In culture, microglia respond to recombinant IL-36γ by expressing molecules involved in neutrophil recruitment, such as Csf3, IL-1β, and Cxcl2. However, mice deficient in either IL-36γ or IL-36R develop similar clinical and histopathological signs of EAE compared to wild-type controls. Conclusion This study identifies IL-36γ as a neutrophil-related cytokine that can potentially activate microglia, but that is only correlative and not contributory in EAE. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0392-7) contains supplementary material, which is available to authorized users.
Collapse
|
48
|
Wang X, Zhao X, Feng C, Weinstein A, Xia R, Wen W, Lv Q, Zuo S, Tang P, Yang X, Chen X, Wang H, Zang S, Stollings L, Denning TL, Jiang J, Fan J, Zhang G, Zhang X, Zhu Y, Storkus W, Lu B. IL-36γ Transforms the Tumor Microenvironment and Promotes Type 1 Lymphocyte-Mediated Antitumor Immune Responses. Cancer Cell 2015; 28:296-306. [PMID: 26321222 PMCID: PMC4573903 DOI: 10.1016/j.ccell.2015.07.014] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 01/11/2015] [Accepted: 07/27/2015] [Indexed: 12/21/2022]
Abstract
Cytokines play a pivotal role in regulating tumor immunogenicity and antitumor immunity. IL-36γ is important for the IL-23/IL-17-dominated inflammation and anti-BCG Th1 immune responses. However, the impact of IL-36γ on tumor immunity is unknown. Here we found that IL-36γ stimulated CD8(+) T cells, NK cells, and γδ T cells synergistically with TCR signaling and/or IL-12. Importantly, IL-36γ exerted profound antitumor effects in vivo and transformed the tumor microenvironment in favor of tumor eradication. Furthermore, IL-36γ strongly increased the efficacy of tumor vaccination. Moreover, IL-36γ expression inversely correlated with the progression of human melanoma and lung cancer. Our study establishes a role of IL-36γ in promoting antitumor immune responses and suggests its potential clinical translation into cancer immunotherapy.
Collapse
Affiliation(s)
- Xuefeng Wang
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Biochemistry and Molecular Biology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China; Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Xin Zhao
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Chao Feng
- Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Aliyah Weinstein
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Dermatology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Rui Xia
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Wen Wen
- Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Quansheng Lv
- Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Shuting Zuo
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Peijun Tang
- Department of Pulmonary Tuberculosis, The Affiliated Hospital for Infectious Diseases of Soochow University, Suzhou 215007, China
| | - Xi Yang
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; School of Medicine, Tsinghua University, Peking 100084, China
| | - Xiaojuan Chen
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Hongrui Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Shayang Zang
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Lindsay Stollings
- Department of Anesthesiology, University of Pittsburgh Medical Center, 3471 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Timothy L Denning
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Jingting Jiang
- Department of Tumor Biotherapy, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh PA 15240, USA
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xueguang Zhang
- Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Yibei Zhu
- Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Walter Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; Department of Dermatology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15261, USA; University of Pittsburgh Cancer Institute, Pittsburgh, PA 15261, USA.
| |
Collapse
|
49
|
Segueni N, Vigne S, Palmer G, Bourigault ML, Olleros ML, Vesin D, Garcia I, Ryffel B, Quesniaux VFJ, Gabay C. Limited Contribution of IL-36 versus IL-1 and TNF Pathways in Host Response to Mycobacterial Infection. PLoS One 2015; 10:e0126058. [PMID: 25950182 PMCID: PMC4423901 DOI: 10.1371/journal.pone.0126058] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 03/28/2015] [Indexed: 11/18/2022] Open
Abstract
IL-36 cytokines are members of the IL-1 family of cytokines that stimulate dendritic cells and T cells leading to enhanced T helper 1 responses in vitro and in vivo; however, their role in host defense has not been fully addressed thus far. The objective of this study was to examine the role of IL-36R signaling in the control of mycobacterial infection, using models of systemic attenuated M. bovis BCG infection and virulent aerogenic M. tuberculosis infection. IL-36γ expression was increased in the lung of M. bovis BCG infected mice. However, IL-36R deficient mice infected with M. bovis BCG showed similar survival and control of the infection as compared to wild-type mice, although their lung pathology and CXCL1 response were transiently different. While highly susceptible TNF-α deficient mice succumbed with overwhelming M. tuberculosis infection, and IL-1RI deficient mice showed intermediate susceptibility, IL-36R-deficient mice controlled the infection, with bacterial burden, lung inflammation and pathology, similar to wild-type controls. Therefore, IL-36R signaling has only limited influence in the control of mycobacterial infection.
Collapse
Affiliation(s)
- Noria Segueni
- CNRS, UMR7355, Orleans, France
- University of Orleans, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Solenne Vigne
- Division of Rheumatology, Department of Internal Medicine Specialties, University Hospitals of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Gaby Palmer
- Division of Rheumatology, Department of Internal Medicine Specialties, University Hospitals of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Marie-Laure Bourigault
- CNRS, UMR7355, Orleans, France
- University of Orleans, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Maria L. Olleros
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Dominique Vesin
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Irene Garcia
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Bernhard Ryffel
- CNRS, UMR7355, Orleans, France
- University of Orleans, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, Cape Town, South Africa
| | - Valérie F. J. Quesniaux
- CNRS, UMR7355, Orleans, France
- University of Orleans, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Cem Gabay
- Division of Rheumatology, Department of Internal Medicine Specialties, University Hospitals of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
- * E-mail:
| |
Collapse
|
50
|
Abstract
The inflammatory status of the tumor microenvironment (TME) has been heavily investigated in recent years. Chemokine- and cytokine-signaling pathways such as CCR7, CXCR5, lymphotoxin, and IL-36, which are involved in the generation of secondary lymphoid organs and effector immune responses, are now recognized as having value both as prognostic factors and as immunomodulatory therapeutics in the context of cancer. Furthermore, when produced in the TME, these mediators have been shown to promote the recruitment of immune cells, including T cells, B cells, dendritic cells (DCs), and other specialized immune cell subsets such as follicular DCs and T follicular helper cells, in association with the formation of "tertiary" lymphoid structures (TLSs) within or adjacent to sites of disease. Although TLSs are composed of a heterogeneous collection of immune cell types, whose composition differs based on cancer subtype, the qualitative presence of TLSs has been shown to represent a biomarker of good prognosis for cancer patients. A comprehensive understanding of the role each of these pathways plays within the TME may support the rational design of future immunotherapies to selectively promote/bolster TLS formation and function, leading to improved clinical outcomes across the vast range of solid cancer types.
Collapse
|