1
|
Lin CI, Wang YW, Su KY, Chuang YH. Interleukin-37 exacerbates liver inflammation and promotes IFN-γ production in NK cells. Int Immunopharmacol 2024; 142:113086. [PMID: 39260304 DOI: 10.1016/j.intimp.2024.113086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/23/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
Interleukin (IL)-37, a unique member of the IL-1 family, is known for its anti-inflammatory properties. However, its effects on immune-mediated liver diseases, such as primary biliary cholangitis (PBC) and acute immune-mediated hepatitis, remain unclear. Using mouse models of autoimmune cholangitis and hepatitis induced by 2-OA-OVA and concanavalin A (Con A) respectively, we introduced the human IL-37 gene via a liver-preferred adeno-associated virus vector (AAV-IL-37) to mice, as mice lack endogenous IL-37. Our findings reveal that IL-37 did not affect autoimmune cholangitis. Surprisingly, IL-37 exacerbated inflammation in Con A-induced hepatitis rather than mitigating it. Mechanistic insights suggest that this exacerbation involves the interferon (IFN)-γ pathway, supported by elevated serum IFN-γ levels in AAV-IL-37-treated Con A mice. Specifically, IL-37 heightened the number of hepatic NK and NKT cells, increased the production of the NK cell chemoattractant CCL5, and elevated the frequency of hepatic NK and NKT cells expressing IFN-γ. Moreover, IL-37 enhanced IFN-γ secretion from NK cells when combined with other proinflammatory cytokines, highlighting its synergistic effect in promoting IFN-γ production. These unexpected outcomes underscore a novel role for IL-37 in exacerbating liver inflammation during immune-mediated liver diseases, implicating its influence on NK cells and the production of IFN-γ by these cells.
Collapse
Affiliation(s)
- Chia-I Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Wen Wang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kang-Yi Su
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan; Genomic and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei, Taiwan
| | - Ya-Hui Chuang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
2
|
Song JH, Hwang B, Lyea Park S, Kim H, Jung S, Choi C, Myung Lee H, Yun SJ, Hyun Choi Y, Cha EJ, Patterson C, Kim WJ, Moon SK. IL-28A/IL-10Rβ axis promotes angiogenesis via eNOS/AKT signaling and AP-1/NF-κB/MMP-2 network by regulating HSP70-1 expression. J Adv Res 2024:S2090-1232(24)00356-4. [PMID: 39127098 DOI: 10.1016/j.jare.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024] Open
Abstract
INTRODUCTION Angiogenesis plays a significant role in the development of tumor progression and inflammatory diseases. The role of IL-28A in angiogenesis and its precise regulatory mechanisms remain rarely elucidated. OBJECTIVES We report the novel regulatory role of IL-28A in physiological angiogenesis. The study aimed to elucidate the regulatory mechanisms involved in IL-28A-mediated angiogenesis and identify key genes associated with IL-28A-induced angiogenic responses. METHODS To know the effect of IL-28A on angiogenesis, HUVECs were applied to perform proliferation, migration, invasion, tube formation, immunoblot, and EMSA. Gene expression changes in HUVECs following IL-28A treatment were analyzed by NGS. The functional role of HSP70-1 and IL-10Rβ in IL-28A-induced angiogenic responses was evaluated using PCR and siRNA knockdown. Animal studies were conducted by aortic ring ex vivo assays, Matrigel plug in vivo assays, and immunochemistry using HSP70-1 knockout and transgenic mice models. The efficacy of IL-28A in angiogenesis was confirmed in a hind-limb ischemia model. RESULTS Autocrine/paracrine actions in HUVECs regulated IL-28A protein expression. Exogenous IL-28A increased the proliferation of HUVECs via eNOS/AKT and ERK1/2 signaling. IL-28A treatment promoted migration, invasion, and capillary tube formation of HUVECs through induction of the AP-1/NF-κB/MMP-2 network, which was associated with eNOS/AKT and ERK1/2 signaling. The efficacy of IL-28A-induced angiogenic potential was confirmed by aortic ring and Matrigel plug assay. HSP70-1 was identified as an IL-28A-mediated angiogenic effector gene using bioinformatics. Knockdown of HSP70-1 abolished angiogenic responses and eNOS/AKT signaling in IL-28A-treated HUVECs. IL-28A-induced microvessel sprouting formation was testified in HSP70-1-deficient and HSP70-1 transgenic mice. Flow recovery in hind-limb ischemia mice was accelerated by IL-28A injection. Finally, ablation of the IL-10Rβ gene impeded the angiogenic responses and eNOS/AKT signaling stimulated by IL-28A in HUVECs. CONCLUSION HSP70-1 drives the progression of angiogenesis by the IL-28A/IL-10Rβ axis via eNOS/AKT signaling and the AP-1/NF-κB/MMP-2 network.
Collapse
Affiliation(s)
- Jun-Hui Song
- Department of Food and Nutrition, Chung-Ang University, Anseong 456-756, Korea
| | - Byungdoo Hwang
- Department of Food and Nutrition, Chung-Ang University, Anseong 456-756, Korea
| | - Sung Lyea Park
- Department of Food and Nutrition, Chung-Ang University, Anseong 456-756, Korea
| | - Hoon Kim
- Department of Food and Nutrition, Chung-Ang University, Anseong 456-756, Korea
| | - Soontag Jung
- Department of Food and Nutrition, Chung-Ang University, Anseong 456-756, Korea
| | - Changsun Choi
- Department of Food and Nutrition, Chung-Ang University, Anseong 456-756, Korea
| | - Hwan Myung Lee
- Department of Cosmetic Science, Hoseo University, Asan-si 31499, Republic of Korea
| | - Seok-Joong Yun
- Personalized Tumor Engineering Research Center, Department of Urology, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dongeui University, Busan 614-052, South Korea
| | - Eun-Jong Cha
- Department of Biomedical Engineering, Chungbuk National University, Cheongju 361-763, Korea
| | - Cam Patterson
- University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Wun-Jae Kim
- Personalized Tumor Engineering Research Center, Department of Urology, Chungbuk National University, Cheongju, Chungbuk 361-763, South Korea; Institute of Urotech, Cheongju, Chungcheongbuk-do 361-763, Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong 456-756, Korea.
| |
Collapse
|
3
|
Ma W, Wu D, Long C, Liu J, Xu L, Zhou L, Dou Q, Ge Y, Zhou C, Jia R. Neutrophil-derived nanovesicles deliver IL-37 to mitigate renal ischemia-reperfusion injury via endothelial cell targeting. J Control Release 2024; 370:66-81. [PMID: 38631490 DOI: 10.1016/j.jconrel.2024.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/09/2024] [Accepted: 04/14/2024] [Indexed: 04/19/2024]
Abstract
Renal ischemia-reperfusion injury (IRI) is one of the most important causes of acute kidney injury (AKI). Interleukin (IL)-37 has been suggested as a novel anti-inflammatory factor for the treatment of IRI, but its application is still limited by its low stability and delivery efficiency. In this study, we reported a novel engineered method to efficiently and easily prepare neutrophil membrane-derived vesicles (N-MVs), which could be utilized as a promising vehicle to deliver IL-37 and avoid the potential side effects of neutrophil-derived natural extracellular vesicles. N-MVs could enhance the stability of IL-37 and targetedly deliver IL-37 to damaged endothelial cells of IRI kidneys via P-selectin glycoprotein ligand-1 (PSGL-1). In vitro and in vivo evidence revealed that N-MVs encapsulated with IL-37 (N-MV@IL-37) could inhibit endothelial cell apoptosis, promote endothelial cell proliferation and angiogenesis, and decrease inflammatory factor production and leukocyte infiltration, thereby ameliorating renal IRI. Our study establishes a promising delivery vehicle for the treatment of renal IRI and other endothelial damage-related diseases.
Collapse
Affiliation(s)
- Wenjie Ma
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Di Wu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Chengcheng Long
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Jingyu Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Luwei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Quanliang Dou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Yuzheng Ge
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Changcheng Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China.
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China.
| |
Collapse
|
4
|
Zhang Y, Zhang Y, Song J, Cheng X, Zhou C, Huang S, Zhao W, Zong Z, Yang L. Targeting the "tumor microenvironment": RNA-binding proteins in the spotlight in colorectal cancer therapy. Int Immunopharmacol 2024; 131:111876. [PMID: 38493688 DOI: 10.1016/j.intimp.2024.111876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/04/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024]
Abstract
Colorectal cancer (CRC) is the third most common cancer and has the second highest mortality rate among cancers. The development of CRC involves both genetic and epigenetic abnormalities, and recent research has focused on exploring the ex-transcriptome, particularly post-transcriptional modifications. RNA-binding proteins (RBPs) are emerging epigenetic regulators that play crucial roles in post-transcriptional events. Dysregulation of RBPs can result in aberrant expression of downstream target genes, thereby affecting the progression of colorectal tumors and the prognosis of patients. Recent studies have shown that RBPs can influence CRC pathogenesis and progression by regulating various components of the tumor microenvironment (TME). Although previous research on RBPs has primarily focused on their direct regulation of colorectal tumor development, their involvement in the remodeling of the TME has not been systematically reported. This review aims to highlight the significant role of RBPs in the intricate interactions within the CRC tumor microenvironment, including tumor immune microenvironment, inflammatory microenvironment, extracellular matrix, tumor vasculature, and CRC cancer stem cells. We also highlight several compounds under investigation for RBP-TME-based treatment of CRC, including small molecule inhibitors such as antisense oligonucleotides (ASOs), siRNAs, agonists, gene manipulation, and tumor vaccines. The insights gained from this review may lead to the development of RBP-based targeted novel therapeutic strategies aimed at modulating the TME, potentially inhibiting the progression and metastasis of CRC.
Collapse
Affiliation(s)
- Yiwei Zhang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 MinDe Road, 330006 Nanchang, China; Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Rd., Nanchang 330006, Jiangxi, China; Queen Mary School, Nanchang University, 330006 Nanchang, China
| | - Yujun Zhang
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 MinDe Road, 330006 Nanchang, China; Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Rd., Nanchang 330006, Jiangxi, China
| | - Jingjing Song
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 MinDe Road, 330006 Nanchang, China; Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Rd., Nanchang 330006, Jiangxi, China; School of Ophthalmology and Optometry of Nanchang University, China
| | - Xifu Cheng
- School of Ophthalmology and Optometry of Nanchang University, China
| | - Chulin Zhou
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Shuo Huang
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Wentao Zhao
- The 3rd Clinical Department of China Medical University, 10159 Shenyang, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 MinDe Road, 330006 Nanchang, China.
| | - Lingling Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Mingde Rd., Nanchang 330006, Jiangxi, China.
| |
Collapse
|
5
|
Wang X, Wang T, Lam E, Alvarez D, Sun Y. Ocular Vascular Diseases: From Retinal Immune Privilege to Inflammation. Int J Mol Sci 2023; 24:12090. [PMID: 37569464 PMCID: PMC10418793 DOI: 10.3390/ijms241512090] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The eye is an immune privileged tissue that insulates the visual system from local and systemic immune provocation to preserve homeostatic functions of highly specialized retinal neural cells. If immune privilege is breached, immune stimuli will invade the eye and subsequently trigger acute inflammatory responses. Local resident microglia become active and release numerous immunological factors to protect the integrity of retinal neural cells. Although acute inflammatory responses are necessary to control and eradicate insults to the eye, chronic inflammation can cause retinal tissue damage and cell dysfunction, leading to ocular disease and vision loss. In this review, we summarized features of immune privilege in the retina and the key inflammatory responses, factors, and intracellular pathways activated when retinal immune privilege fails, as well as a highlight of the recent clinical and research advances in ocular immunity and ocular vascular diseases including retinopathy of prematurity, age-related macular degeneration, and diabetic retinopathy.
Collapse
Affiliation(s)
- Xudong Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| | - Tianxi Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| | - Enton Lam
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| | - David Alvarez
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| |
Collapse
|
6
|
Mariotti FR, Supino D, Landolina N, Garlanda C, Mantovani A, Moretta L, Maggi E. IL-1R8: A molecular brake of anti-tumor and anti-viral activity of NK cells and ILC. Semin Immunol 2023; 66:101712. [PMID: 36753974 DOI: 10.1016/j.smim.2023.101712] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 02/07/2023]
Abstract
Interleukin-1 receptor family members (ILRs) and Toll-Like Receptors (TLRs) play pivotal role in immunity and inflammation and are expressed by most cell types including cells of both the innate and adaptive immune system. In this context, IL-1 superfamily members are also important players in regulating function and differentiation of adaptive and innate lymphoid cells. This system is tightly regulated in order to avoid uncontrolled activation, which may lead to detrimental inflammation contributing to autoimmune or allergic responses. IL-1R8 (also known as TIR8 or SIGIRR) is a member of the IL-1R family that acts as a negative regulator dampening ILR and TLR signaling and as a co-receptor for human IL-37. Human and mouse NK cells, that are key players in immune surveillance of tumors and infections, express high level of IL-1R8. In this review, we will summarize our current understanding on the structure, expression and function of IL-1R8 and we will also discuss the emerging role of IL-1R8 as an important checkpoint regulating NK cells function in pathological conditions including cancer and viral infections.
Collapse
Affiliation(s)
- Francesca R Mariotti
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | | | - Nadine Landolina
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Cecilia Garlanda
- IRCCS, Humanitas Research Hospital, 20089 Rozzano, Italy; Department of Biomedical Science, Humanitas University, 20072 Pieve Emanuele, Italy
| | - Alberto Mantovani
- IRCCS, Humanitas Research Hospital, 20089 Rozzano, Italy; Department of Biomedical Science, Humanitas University, 20072 Pieve Emanuele, Italy; The William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Enrico Maggi
- Translational Immunology Unit, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy.
| |
Collapse
|
7
|
Green EA, Garrick SP, Peterson B, Berger PJ, Galinsky R, Hunt RW, Cho SX, Bourke JE, Nold MF, Nold-Petry CA. The Role of the Interleukin-1 Family in Complications of Prematurity. Int J Mol Sci 2023; 24:2795. [PMID: 36769133 PMCID: PMC9918069 DOI: 10.3390/ijms24032795] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/20/2023] [Accepted: 01/22/2023] [Indexed: 02/05/2023] Open
Abstract
Preterm birth is a major contributor to neonatal morbidity and mortality. Complications of prematurity such as bronchopulmonary dysplasia (BPD, affecting the lung), pulmonary hypertension associated with BPD (BPD-PH, heart), white matter injury (WMI, brain), retinopathy of prematurity (ROP, eyes), necrotizing enterocolitis (NEC, gut) and sepsis are among the major causes of long-term morbidity in infants born prematurely. Though the origins are multifactorial, inflammation and in particular the imbalance of pro- and anti-inflammatory mediators is now recognized as a key driver of the pathophysiology underlying these illnesses. Here, we review the involvement of the interleukin (IL)-1 family in perinatal inflammation and its clinical implications, with a focus on the potential of these cytokines as therapeutic targets for the development of safe and effective treatments for early life inflammatory diseases.
Collapse
Affiliation(s)
- Elys A. Green
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC 3168, Australia
| | - Steven P. Garrick
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Briana Peterson
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Philip J. Berger
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Robert Galinsky
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC 3168, Australia
| | - Rod W. Hunt
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC 3168, Australia
| | - Steven X. Cho
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Jane E. Bourke
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3168, Australia
| | - Marcel F. Nold
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC 3168, Australia
| | - Claudia A. Nold-Petry
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| |
Collapse
|
8
|
Cui G, Liu H, Laugsand JB. Endothelial cells-directed angiogenesis in colorectal cancer: Interleukin as the mediator and pharmacological target. Int Immunopharmacol 2023; 114:109525. [PMID: 36508917 DOI: 10.1016/j.intimp.2022.109525] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/22/2022] [Accepted: 11/27/2022] [Indexed: 12/14/2022]
Abstract
Enhanced angiogenesis is a cancer hallmark and critical for colorectal cancer (CRC) invasion and metastasis. Upon exposure to proangiogenic factors, therefore, targeting tumor-associated proangiogenic factors/receptors hold great promise as a therapeutic modality to treat CRC, particularly metastatic CRC. Accumulating evidence from numerous studies suggests that tumor endothelial cells (ECs) are not only the target of proangiogenic factors, but also function as the cellular source of proangiogenic factors. Studies showed that ECs can produce different proangiogenic factors to participate in the regulation of angiogenesis process, in which ECs-derived interleukins (ILs) show a potential stimulatory effect on angiogenesis via either an direct action on their receptors expressed on progenitor of ECs or an indirect way through enhanced production of other proangiogenic factors. Although a great deal of attention is given to the effects of tumor-derived and immune cell-derived ILs, few studies describe the potential effects of vascular ECs-derived ILs on the tumor angiogenesis process. This review provides an updated summary of available information on proangiogenic ILs, such as IL-1, IL-6, IL-8, IL-17, IL-22, IL-33, IL-34, and IL-37, released by microvascular ECs as potential drivers of the tumor angiogenesis process and discusses their potential as a novel candidate for antiangiogenic target for the treatment of CRC patients.
Collapse
Affiliation(s)
- Guanglin Cui
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Faculty of Health Science, Nord University, Campus Levanger, Norway.
| | - Hanzhe Liu
- School of Stomatology, Wuhan University, Wuhan, China.
| | | |
Collapse
|
9
|
Zhang J, Yan H, Li J, Li B. Aqueous Humor Factors' Predictive Effects in Treating Refractor Macular Edema: An Overview. J Interferon Cytokine Res 2022; 42:515-524. [PMID: 36036998 DOI: 10.1089/jir.2022.0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The refractory macular edema (RME) seriously affects the patient's vision due to its repetition and ineffective drug response. RME is mainly related to the inflammatory pathway and angiogenesis pathway. At present, microglia activation and angiogenesis have also been widely focused on. With the promotion of the concept of precision diagnosis and treatment, intraocular fluid is becoming a popular evidence-based method. The detection and evaluation of aqueous humor factors can provide more accurate evidences and guidance for the treatment of RME. The purpose of this article is to review the treatment prediction and assessment progress of aqueous humor cytokines for the RME, giving evidence to provide a basis for expanding the diagnosis and treatment ideas of RME and guiding the development of personalized medical treatment.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Ophthalmology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hui Yan
- Department of Ophthalmology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jing Li
- Department of Ophthalmology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bing Li
- Department of Ophthalmology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
10
|
Elsaid DS, Elbedewy TAH, Soliman NA, Shalaby KA, Abdel-Hamid Haroun R. Interleukin-37, vascular endothelial growth factor A, and transforming growth factor-β1: promising biomarkers in primary immune thrombocytopenia. Expert Rev Hematol 2022; 15:757-768. [PMID: 35815383 DOI: 10.1080/17474086.2022.2099832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Immune thrombocytopenic purpura (ITP) is an acquired autoimmune hematologic disorder with heterogeneous bleeding manifestations. Many biomarkers such as interleukin-37 (IL-37), vascular endothelial growth factor A (VEGFA), and transforming growth factor-β1 (TGFß1) have a role in immunity, inflammation, and megakaryopoiesis. METHODS In the present study, immunoassay of interleukin-37 as well as the gene expression of vascular endothelial growth factor A and transforming growth factor-β1 were done in 60 primary ITP patients, 60 thrombocytopenia patients, and 60 healthy volunteers. RESULTS Increased IL-37 level and down regulation of VEGFA and TGFß1gene expression were detected in primary ITP patients when compared with other groups. A negative correlation was observed between IL-37 and platelet count. However, a positive correlation was observed between VEGFA and TGFß1 levels and platelet count. CONCLUSION Current results suggested that interleukin-37, vascular endothelial growth factor A, and transforming growth factor-β may be promising indicators in the diagnosis of ITP and detection of disease severity with inexpensive and cost-effectiveness compared to the benefits.
Collapse
Affiliation(s)
- Dina Samir Elsaid
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | | | - Nema Ali Soliman
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Kamal Ali Shalaby
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | | |
Collapse
|
11
|
Wu P, Zhou J, Wu Y, Zhao L. The emerging role of Interleukin 37 in bone homeostasis and inflammatory bone diseases. Int Immunopharmacol 2021; 98:107803. [PMID: 34091255 DOI: 10.1016/j.intimp.2021.107803] [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: 03/23/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 02/05/2023]
Abstract
Interleukin 37 (IL-37) is a newly identified cytokine that belongs to the IL-1 family. Unlike other members of the IL-1 family, it has been demonstrated that IL-37 possesses anti-inflammatory characteristics in both innate and acquired immune responses. Recently, significant progress has been made in understanding the role of IL-37 in inflammatory signaling pathways. Meanwhile, IL-37 has also attracted more and more attention in bone homeostasis and inflammatory bone diseases. The latest studies have revealed that IL-37 palys an essential role in the regulation of osteoclastogenesis and osteoblastogenesis. The levels of IL-37 are abnormal in patients with inflammatory bone diseases such as rheumatoid arthritis (RA), osteoarthritis (OA), ankylosing spondylitis (AS), and periodontitis. In addition, in vivo studies have further confirmed that recombinant IL-37 treatment displayed therapeutic potential in these diseases. The present review article aims to provide an overview describing the biological functions of IL-37 in bone homeostasis and inflammatory bone diseases, thus shedding new light on a novel therapeutic strategy in the future.
Collapse
Affiliation(s)
- Peiyao Wu
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jieyu Zhou
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yafei Wu
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Lei Zhao
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
12
|
He Y, Xiong T, Guo F, Du Z, Fan Y, Sun H, Feng Z, Zhang G. Interleukin-37b inhibits the growth of murine endometriosis-like lesions by regulating proliferation, invasion, angiogenesis and inflammation. Mol Hum Reprod 2021; 26:240-255. [PMID: 32119739 DOI: 10.1093/molehr/gaaa014] [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: 12/01/2019] [Revised: 01/28/2020] [Accepted: 02/06/2020] [Indexed: 12/17/2022] Open
Abstract
Endometriosis is a gynecological disease with abnormal expression of interleukin (IL)-37 which can suppress inflammation and the immune system. Here we investigated the role of the IL-37b splice variant in endometriosis in vivo and in vitro. In a murine model of endometriosis, in vivo administration of IL-37b significantly inhibited the development of lesions judged by the number (P = 0.0213), size (P = 0.0130) and weight (P = 0.0152) of lesions. IL-37b had no effect on the early stage of lesion formation, however administration in the growth stage of lesions decreased the number (P = 0.0158), size (P = 0.0158) and weight (P = 0.0258) of lesions compared with PBS control, an effect that was not reversed by macrophage depletion. Expressions of inflammatory factors, matrix metalloproteinases and vascular endothelial growth factor-A mRNA/protein were significantly inhibited in ectopic lesions following IL-37b administration, and in uterine segments treated in vitro. In vitro treatment of uterine segments with IL-37b inhibited phosphorylation of Akt and Erk1/2 in uterine segments. Isolated mouse endometrial stromal treated with IL-37b and transfected with pIL-37b plasmid got suppressed cell proliferation, invasion, angiogenesis and the expression of inflammatory factors. In addition, transfection with pIL-37b significantly decreased the phosphorylation of Akt and Erk1/2. IL-37b also inhibited proliferation and the expression of inflammatory and angiogenesis factors in epithelial cell line RL95-2. These findings suggest that IL-37b may inhibit the growth of lesions by regulating proliferation, invasion, angiogenesis and inflammation through Akt and Erk1/2 signaling pathway.
Collapse
Affiliation(s)
- Yongpei He
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, The people's Republic of China
| | - Ting Xiong
- Department of Gynaecology and Obstetrics, Reproductive Medical center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, The People's Republic of China
| | - Fang Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, The people's Republic of China
| | - Zhenzhen Du
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, The people's Republic of China
| | - Yixian Fan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, The people's Republic of China
| | - Huanhuan Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, The people's Republic of China
| | - Zuohua Feng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, The people's Republic of China
| | - Guimei Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, The people's Republic of China
| |
Collapse
|
13
|
Lee J, Jin YJ, Lee MS, Kim YM, Lee H. Macrophage inhibitory cytokine-1 promotes angiogenesis by eliciting the GFRAL-mediated endothelial cell signaling. J Cell Physiol 2021; 236:4008-4023. [PMID: 33151561 DOI: 10.1002/jcp.30144] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 11/09/2022]
Abstract
Macrophage inhibitory cytokine-1 (MIC-1) is a cytokine with pleotropic actions and its expression is markedly increased by inflammation and cardiac injury and in cancers. In particular, MIC-1 production after cardiac ischemia injury is associated with enhanced cardiac angiogenesis as well as myocardial protection. However, it remains uncertain whether MIC-1 itself has proangiogenic activity. In this study, we tried to determine the precise role of MIC-1 in physiological and pathological angiogenesis. Human microvessel endothelial cells responded to MIC-1 with enhanced angiogenic behaviors. Employing various angiogenesis assays, MIC-1 was found to promote vessel formation and development with a potency similar to that of vascular endothelial growth factor (VEGF). MIC-1 transgenic (Tg) mice also displayed enhanced neovascularization in both developing embryos and neonatal mouse retinas, compared with wild-type mice. Furthermore, endothelial cells (ECs) isolated from MIC-1 Tg mouse lung exhibited higher angiogenic potential than ECs from wild-type lung. MIC-1-induced angiogenesis was also observed in the recovery or healing processes of injuries such as hindlimb ischemia and skin wounds in mice. However, unlike VEGF, MIC-1 induced neither endothelial inflammation nor increased vascular permeability. In ECs, the MIC-1 signal exerted proangiogenic actions via the MEK/extracellular signal-regulated kinase- and phosphatidylinositol 3-kinase/Akt-dependent pathways. Notably, these MIC-1 signaling events in ECs were abrogated by small interfering RNA-mediated knockdown of GFRAL, suggesting that GFRAL is an EC receptor for MIC-1. In summary, we here show a novel role of MIC-1 as a potent EC activator, which promotes both normal and injury-related angiogenesis.
Collapse
Affiliation(s)
- Jaeseob Lee
- Department of Biological Sciences, Kangwon National University, Chunchon, Kangwon-do, South Korea
| | - Young-June Jin
- Department of Biological Sciences, Kangwon National University, Chunchon, Kangwon-do, South Korea
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Moon-Sung Lee
- Department of Biological Sciences, Kangwon National University, Chunchon, Kangwon-do, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, Kangwon National University, Chunchon, Kangwon-do, South Korea
| | - Hansoo Lee
- Department of Biological Sciences, Kangwon National University, Chunchon, Kangwon-do, South Korea
| |
Collapse
|
14
|
Serum Interleukin-37 Increases in Patients after Ischemic Stroke and Is Associated with Stroke Recurrence. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5546991. [PMID: 33953828 PMCID: PMC8057878 DOI: 10.1155/2021/5546991] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/23/2021] [Accepted: 04/04/2021] [Indexed: 01/11/2023]
Abstract
Background This study seeks to assess interleukin-37 (IL-37) serum level in acute ischemic stroke and the value of predicting 3-month stroke recurrence and functional outcome in acute ischemic stroke. Methods From January 1, 2018, to June 30, 2019, all consecutive first-ever acute ischemic stroke patients from our hospital, China, were included. Serum samples, clinical information, and stroke severity (defined by the National Institute of Health stroke scale (NIHSS) score) were collected at baseline. Serum IL-37 level was measured by the enzyme-linked immunosorbent assay (ELISA) method. Functional impairment (defined by the modified Rankin scale (mRS)) and recurrent stroke were assessed 3 months after admission. The relation of IL-37 with either clinical severity at baseline, unfavorable functional outcome, or stroke recurrence at follow-up was evaluated by logistic regression analysis, and the results were presented as odds ratios (OR) with 95% confidence intervals (CI). Results Three hundred and ten stroke patients were included. The median IL-37 serum level in those patients was 344.1 pg/ml (interquartile range (IQR), 284.4-405.3 vs. control cases: 122.3 pg/ml (IQR, 104.4-1444.0); P < 0.001). At 3 months, a total of 36 (11.6%) patients had a stroke recurrence. IL-37 serum levels in those patients were higher than in those patients without stroke recurrence (417.0 pg/ml (IQR, 359.3-436.1) vs. 333.3 pg/ml (279.0-391.0)). In a logistic model adjusted for other factors, IL-37 in the highest quartile (>405.3 pg/ml) was still associated with recurrent stroke (OR = 3.32; 95%CI = 2.03–6.13; P < 0.001). IL-37 could promote the NIHSS score (area under the curve (AUC) of the IL-37/NIHSS, 0.75; 95% CI, 0.67–0.83; P < 0.001), corresponding to a difference of 0.085 (0.005). Serum IL-37 increases in patients with poor outcome, and an IL-37 in the highest quartile is related to poor outcome (OR = 4.85; 95%CI = 3.11 − 8.22; P < 0.001). Conclusion Serum IL-37 increased in patients after ischemic stroke and was associated with stroke recurrence events and poor stroke outcomes. Large randomized controlled trials should be carried out to confirm whether IL-37 lowering treatment improves stroke prognosis.
Collapse
|
15
|
Jiang X, Wang J, Deng X, Xiong F, Zhang S, Gong Z, Li X, Cao K, Deng H, He Y, Liao Q, Xiang B, Zhou M, Guo C, Zeng Z, Li G, Li X, Xiong W. The role of microenvironment in tumor angiogenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:204. [PMID: 32993787 PMCID: PMC7526376 DOI: 10.1186/s13046-020-01709-5] [Citation(s) in RCA: 338] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022]
Abstract
Tumor angiogenesis is necessary for the continued survival and development of tumor cells, and plays an important role in their growth, invasion, and metastasis. The tumor microenvironment—composed of tumor cells, surrounding cells, and secreted cytokines—provides a conducive environment for the growth and survival of tumors. Different components of the tumor microenvironment can regulate tumor development. In this review, we have discussed the regulatory role of the microenvironment in tumor angiogenesis. High expression of angiogenic factors and inflammatory cytokines in the tumor microenvironment, as well as hypoxia, are presumed to be the reasons for poor therapeutic efficacy of current anti-angiogenic drugs. A combination of anti-angiogenic drugs and antitumor inflammatory drugs or hypoxia inhibitors might improve the therapeutic outcome.
Collapse
Affiliation(s)
- Xianjie Jiang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Jie Wang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Xiangying Deng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Fang Xiong
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Shanshan Zhang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ke Cao
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hao Deng
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yi He
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Bo Xiang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Ming Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Can Guo
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Xiaoling Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China.
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China.
| |
Collapse
|
16
|
Mei Y, Zhu Y, Teo HY, Liu Y, Song Y, Lim HY, Binte Hanafi Z, Angeli V, Liu H. The indirect antiangiogenic effect of IL-37 in the tumor microenvironment. J Leukoc Biol 2020; 107:783-796. [PMID: 32125036 DOI: 10.1002/jlb.3ma0220-207rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 01/28/2023] Open
Abstract
IL-37, a newly identified IL-1 family cytokine, has been shown to play an important role in inflammatory diseases, autoimmune diseases, and carcinogenesis. IL-37 has been suggested to suppress tumoral angiogenesis, whereas some publications showed that IL-37 promoted angiogenesis through TGF-β signaling in both physiologic and pathologic conditions. Therefore, the function of IL-37 in tumoral angiogenesis is not clear and the underlying mechanism is not known. In this current study, we investigated the direct role of IL-37 on endothelial cells, as well as its indirect effect on angiogenesis through functioning on tumor cells both in vitro and in vivo. We found that IL-37 treatment directly promoted HUVEC migration and tubule formation, indicating IL-37 as a proangiogenic factor. Surprisingly, the supernatants from IL-37 overexpressing tumor cell line promoted HUVEC apoptosis and inhibited its migration and tubule formation. Furthermore, we demonstrated that IL-37 suppressed tumor angiogenesis in a murine orthotopic hepatocellular carcinoma model, suggesting its dominant antiangiogenesis role in vivo. Moreover, microarray and qPCR analysis demonstrated that IL-37 reduced the expressions of proangiogenic factors and increased the expressions of antiangiogenic factors by tumor cells. Matrix metalloproteinase (MMP)2 expression was significantly decreased by IL-37 in both cell lines and murine tumor models. MMP9 and vascular endothelial growth factor expressions were also reduced in murine tumors overexpressing IL-37, as well as in cell lines overexpressing IL-37 under hypoxic conditions. In conclusion, although IL-37 could exert direct proangiogenic effects on endothelial cells, it plays an antiangiogenic role via modulating proangiogenic and antiangiogenic factor expressions by tumor cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Yu Mei
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Ying Zhu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Huey Yee Teo
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Yonghao Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Yuan Song
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Hwee Ying Lim
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Zuhairah Binte Hanafi
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Veronique Angeli
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
17
|
Pan Y, Wen X, Hao D, Wang Y, Wang L, He G, Jiang X. The role of IL-37 in skin and connective tissue diseases. Biomed Pharmacother 2019; 122:109705. [PMID: 31918276 DOI: 10.1016/j.biopha.2019.109705] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/12/2019] [Accepted: 11/24/2019] [Indexed: 02/05/2023] Open
Abstract
IL-37 was discovered as an anti-inflammatory and immunosuppressive cytokine of the IL-1 family. Significant advancements in the understanding of signaling pathways associated with IL-37 have been made in recent years. IL-37 binds to IL-18R and recruits IL-1R8 to form the IL-37/IL-1R8/IL-18Rα complex. Capase-1 plays a key role in the nuclear transduction of IL-37 signal, processing precursor IL-37 into the mature isoform, and interacting with Smad3. IL-37 exerts its role by activating anti-inflammation pathways including AMPK, PTEN, Mer, STAT3 and p62, and promoting tolerogenic dendritic cells and Tregs. In addition, IL-37 inhibits pro-inflammatory cytokines such as IL-1, IL-6, IL-8, IL-17, IL-23, TNF-α, and IFN-γ, and suppresses Fyn, MAPK, TAK1, NFκB, and mTOR signaling. The final effects of IL-37 depend on the interaction among IL-18R, IL-1R8, IL-37 and IL-18BP. Previous studies have deciphered the role of IL-37 in the development and pathogenesis of autoimmune diseases, chronic infections and cancer. In this review, we discuss the role of IL-37 in psoriasis, atopic dermatitis, Behcet's diseases, systemic lupus erythematosus, and other skin and connective tissue diseases.
Collapse
Affiliation(s)
- Yu Pan
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xiang Wen
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Dan Hao
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yujia Wang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Lian Wang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Gu He
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, PR China.
| | - Xian Jiang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
18
|
Role of Interleukin 37 as a Novel Proangiogenic Factor in Juvenile Idiopathic Arthritis. J Clin Rheumatol 2019; 25:85-90. [PMID: 29683837 DOI: 10.1097/rhu.0000000000000779] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The aim of this study was to investigate interleukin 37 (IL-37) levels in the serum and synovial fluid of patients with juvenile idiopathic arthritis (JIA), its expression in peripheral blood mononuclear cells, and correlation with disease activity and angiogenesis. METHODS Seventy JIA patients and 50 control subjects were examined. The Juvenile Arthritis Disease Activity Score in 27 joints (JADAS-27) was calculated. Immunoassays were used to measure the serum and synovial fluid levels of IL-37, vascular endothelial growth factor (VEGF), soluble VEGF receptor 1 (sVEGF-R1), and sVEGF-R2. Relative expression of IL-37 mRNA in peripheral blood mononuclear cells and the power Doppler ultrasound score of the affected joint were measured. RESULTS Patients with JIA were subdivided as 20 systemic-onset, 20 polyarticular, and 30 oligoarticular (10 persistent, 20 extended) cases. Serum levels of IL-37, VEGF, VEGF-R1, and VEGF-R2 and relative IL-37 mRNA expression were significantly higher in JIA patients when compared with the control subjects (p < 0.001). These concentrations were significantly higher in systemic-onset JIA compared with those in polyarticular and oligoarticular JIA, and in polyarticular JIA when compared with oligoarticular JIA (p < 0.001). Serum, synovial, and mRNA expression levels of IL-37 were positively correlated with C-reactive protein, erythrocyte sedimentation rate, Juvenile Arthritis Disease Activity Score in 27 joints, power Doppler ultrasound score (p < 0.001), and the serum and synovial VEGF and VEGF-RI and -R2 levels (p < 0.05). CONCLUSIONS Our results demonstrate that IL-37 levels and mRNA expression were significantly increased in JIA patients, and their levels were positively correlated with disease activity and markers of angiogenesis (VEGF and VEGF receptors), suggesting that IL-37 may be correlated with angiogenesis.
Collapse
|
19
|
Extracellular IL-37 promotes osteogenic differentiation of human bone marrow mesenchymal stem cells via activation of the PI3K/AKT signaling pathway. Cell Death Dis 2019; 10:753. [PMID: 31582734 PMCID: PMC6776644 DOI: 10.1038/s41419-019-1904-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 07/24/2019] [Accepted: 08/11/2019] [Indexed: 12/11/2022]
Abstract
Interleukin (IL)-37, a pivotal anti-inflammatory cytokine and a fundamental inhibitor of innate immunity, has recently been shown to be abnormally expressed in several autoimmune-related orthopedic diseases, including rheumatoid arthritis, ankylosing spondylitis, and osteoporosis. However, the role of IL-37 during osteogenic differentiation of mesenchymal stem cells (MSCs) remains largely unknown. In this study, extracellular IL-37 significantly increased osteoblast-specific gene expression, the number of mineral deposits, and alkaline phosphatase activity of MSCs. Moreover, a signaling pathway was activated in the presence of IL-37. The enhanced osteogenic differentiation of MSCs due to supplementation of IL-37 was partially rescued by the presence of a PI3K/AKT signaling inhibitor. Using a rat calvarial bone defect model, IL-37 significantly improved bone healing. Collectively, these findings indicate that extracellular IL-37 enhanced osteogenesis of MSCs, at least in part by activation of the PI3K/AKT signaling pathway.
Collapse
|
20
|
Liu T, Liu J, Lin Y, Que B, Chang C, Zhang J, Liang Z, Gao X, Liu S, Liu L, Li D, Ji Q. IL-37 inhibits the maturation of dendritic cells through the IL-1R8-TLR4-NF-κB pathway. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1338-1349. [DOI: 10.1016/j.bbalip.2019.05.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 12/31/2022]
|
21
|
Wooff Y, Man SM, Aggio-Bruce R, Natoli R, Fernando N. IL-1 Family Members Mediate Cell Death, Inflammation and Angiogenesis in Retinal Degenerative Diseases. Front Immunol 2019; 10:1618. [PMID: 31379825 PMCID: PMC6646526 DOI: 10.3389/fimmu.2019.01618] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/28/2019] [Indexed: 12/22/2022] Open
Abstract
Inflammation underpins and contributes to the pathogenesis of many retinal degenerative diseases. The recruitment and activation of both resident microglia and recruited macrophages, as well as the production of cytokines, are key contributing factors for progressive cell death in these diseases. In particular, the interleukin 1 (IL-1) family consisting of both pro- and anti-inflammatory cytokines has been shown to be pivotal in the mediation of innate immunity and contribute directly to a number of retinal degenerations, including Age-Related Macular Degeneration (AMD), diabetic retinopathy, retinitis pigmentosa, glaucoma, and retinopathy of prematurity (ROP). In this review, we will discuss the role of IL-1 family members and inflammasome signaling in retinal degenerative diseases, piecing together their contribution to retinal disease pathology, and identifying areas of research expansion required to further elucidate their function in the retina.
Collapse
Affiliation(s)
- Yvette Wooff
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia.,ANU Medical School, The Australian National University, Canberra, ACT, Australia
| | - Si Ming Man
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Riemke Aggio-Bruce
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Riccardo Natoli
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia.,ANU Medical School, The Australian National University, Canberra, ACT, Australia
| | - Nilisha Fernando
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
22
|
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: 160] [Impact Index Per Article: 32.0] [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
|
23
|
Baker KJ, Houston A, Brint E. IL-1 Family Members in Cancer; Two Sides to Every Story. Front Immunol 2019; 10:1197. [PMID: 31231372 PMCID: PMC6567883 DOI: 10.3389/fimmu.2019.01197] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/13/2019] [Indexed: 12/22/2022] Open
Abstract
The IL-1 family of cytokines currently comprises of seven ligands with pro-inflammatory activity (IL-1α and IL-1β, IL-18, IL-33, IL-36α, IL-36β, IL-36γ) as well as two ligands with anti-inflammatory activity (IL-37, IL-38). These cytokines are known to play a key role in modulating both the innate and adaptive immunes response, with dysregulation linked to a variety of autoimmune and inflammatory diseases. Given the increasing appreciation of the link between inflammation and cancer, the role of several members of this family in the pathogenesis of cancer has been extensively investigated. In this review, we highlight both the pro- and anti-tumorigenic effects identified for almost all members of this family, and explore potential underlying mechanisms accounting for these divergent effects. Such dual functions need to be carefully assessed when developing therapeutic intervention strategies targeting these cytokines in cancer.
Collapse
Affiliation(s)
- Kevin J Baker
- Department of Pathology, University College Cork, Cork, Ireland.,Department of Medicine, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Aileen Houston
- Department of Medicine, University College Cork, Cork, Ireland.,CancerResearch@UCC, University College Cork, Cork, Ireland
| | - Elizabeth Brint
- Department of Pathology, University College Cork, Cork, Ireland.,CancerResearch@UCC, University College Cork, Cork, Ireland
| |
Collapse
|
24
|
van Geffen EW, van Caam APM, Schreurs W, van de Loo FA, van Lent PLEM, Koenders MI, Thudium CS, Bay-Jensen AC, Blaney Davidson EN, van der Kraan PM. IL-37 diminishes proteoglycan loss in human OA cartilage: donor-specific link between IL-37 and MMP-3. Osteoarthritis Cartilage 2019; 27:148-157. [PMID: 30201492 DOI: 10.1016/j.joca.2018.08.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 07/23/2018] [Accepted: 08/29/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE A hallmark of osteoarthritis (OA) is degradation of articular cartilage proteoglycans. In isolated human OA chondrocytes, the anti-inflammatory cytokine Interleukin-37 (IL-37) lowers the expression of the proteolytic MMP and ADAMTS enzymes, which mediate this degradation. Therefore, we investigated if IL-37 protects against proteoglycan loss in freshly obtained human OA explants. MATERIAL AND METHODS Human OA cartilage explants were incubated with IL-37. Release of sulphated proteoglycans (sGAGs) was measured with the dimethylmethylene-blue assay. Production and degradation of newly synthesized proteoglycans was measured using 35S-sulphate. Proteoglycan and proteolytic enzyme expression were analyzed by qPCR and Western Blot. Proteolytic activity was determined by measuring MMP- and ADAMTS-generated aggrecan neo-epitopes with ELISA and by using MMP-3-, MMP-13- or ADAMTS-5-inhibitors. RESULTS Over time, a linear release of sGAGs from OA cartilage was measured. IL-37 reduced this release by 87 μg/ml (24%) 95%CI [21.04-141.4]. IL-37 did not affect 35S-sulphate incorporation or proteoglycan gene expression. In contrast, IL-37 reduced loss of 35S-sulphate labeled GAGs and reduced MMP-3 protein expression, indicating that IL-37 inhibits proteoglycan degradation. Remarkably, we observed two groups of patients; one group in which MMP-3-inhibition lowered sGAG release, and one group in which ADAMTS5-inhibition had this effect. Remarkably, IL-37 was only functional in the group of patients that responded to MMP-3-inhibition. CONCLUSION We identified a relationship between IL-37 and reduced sGAG loss in OA cartilage. Most likely, this effect is mediated by inhibition of MMP-3 expression. These results suggest that IL-37 could be applied as therapy in a subgroup of OA patients, in which cartilage degradation is mediated by MMP-3.
Collapse
Affiliation(s)
- E W van Geffen
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A P M van Caam
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W Schreurs
- Department of Orthopaedics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - F A van de Loo
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - P L E M van Lent
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M I Koenders
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - C S Thudium
- Department of Rheumatology, Nordic Bioscience, Copenhagen, Denmark
| | - A C Bay-Jensen
- Department of Rheumatology, Nordic Bioscience, Copenhagen, Denmark
| | - E N Blaney Davidson
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - P M van der Kraan
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
25
|
Mei Y, Liu H. IL-37: An anti-inflammatory cytokine with antitumor functions. Cancer Rep (Hoboken) 2018; 2:e1151. [PMID: 32935478 DOI: 10.1002/cnr2.1151] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND IL-37 is a newly identified IL-1 family cytokine. Unlike other members in IL-1 family, IL-37 has been demonstrated to be an anti-inflammatory cytokine in many inflammatory and autoimmune diseases. IL-37 is regarded as a dual-function cytokine as both the extracellular and intracellular IL-37 are biologically functional. Extracellular IL-37 can bind to IL-18Rα and IL-1R8 to form a triple complex, regulating the downstream STAT3 and PTEN signaling. Intracellular IL-37 can interact with Smad3, translocate into nucleus, and regulate downstream target gene expressions. Recently, the role of IL-37 in tumor development has been extensively studied. RECENT FINDINGS IL-37 has been found to play an antitumor role in various types of tumors, such as non-small cell lung cancer, hepatocellular carcinoma, and renal cell carcinoma. Many mechanism studies have been carried out to elaborate the possible effects of IL-37 on tumor growth, immune responses, and tumor angiogenesis. More importantly, the function of IL-37 may be dependent on its concentration and receptor expression. It can form dimers at high concentrations to be inactivated, thus inhibiting its anti-inflammatory function. We focused on the role of IL-37 in various tumor types and provided the hypothesis regarding the underlying mechanisms. CONCLUSION IL-37 may affect tumor development through multiple mechanisms: (1) IL-37 directly influences tumor cell viability; (2) IL-37 regulates the immune response to promote the antitumor immunity; and (3) IL-37 suppresses tumor angiogenesis in the tumor microenvironment. Future studies are warranted to further investigate the mechanisms of these multifaceted functions of IL-37 in animal models and cancer patients.
Collapse
Affiliation(s)
- Yu Mei
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, National University of Singapore, Singapore
| |
Collapse
|
26
|
Tang R, Yi J, Yang J, Chen Y, Luo W, Dong S, Fei J. Interleukin-37 inhibits osteoclastogenesis and alleviates inflammatory bone destruction. J Cell Physiol 2018; 234:7645-7658. [PMID: 30414292 PMCID: PMC6587950 DOI: 10.1002/jcp.27526] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/10/2018] [Indexed: 12/30/2022]
Abstract
Excessive osteoclast formation is one of the important pathological features of inflammatory bone destruction. Interleukin‐37 (IL‐37) is an anti‐inflammatory agent that is present throughout the body, but it displays low physiological retention. In our study, high levels of the IL‐37 protein were detected in clinical specimens from patients with bone infections. However, the impact of IL‐37 on osteoclast formation remains unclear. Next, IL‐37 alleviated the inflammatory bone destruction in the mouse in vivo. We used receptor activator of nuclear factor‐κB ligand and lipopolysaccharide to trigger osteoclastogenesis under physiological and pathological conditions to observe the role of IL‐37 in this process and explore the potential mechanism of this phenomenon. In both induction models, IL‐37 exerted inhibitory effects on osteoclast differentiation and bone resorption. Furthermore, IL‐37 decreased the phosphorylation of inhibitor of κBα and p65 and the expression of nuclear factor of activated T cells c1, while the dimerization inhibitor of myeloid differentiation factor 88 reversed the effects. These data provide evidence that IL‐37 modulates osteoclastogenesis and a theoretical basis for the clinical application of IL‐37 as a treatment for bone loss–related diseases.
Collapse
Affiliation(s)
- Ruohui Tang
- Center of Trauma of Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jin Yi
- Center of Trauma of Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jing Yang
- Center of Trauma of Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yueqi Chen
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wei Luo
- Department of Osteological, Guizhou Province People's Hospital, Guiyang, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China
| | - Jun Fei
- Center of Trauma of Daping Hospital, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China
| |
Collapse
|
27
|
IL-37 Expression Reduces Lean Body Mass in Mice by Reducing Food Intake. Int J Mol Sci 2018; 19:ijms19082264. [PMID: 30072596 PMCID: PMC6121375 DOI: 10.3390/ijms19082264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022] Open
Abstract
The human cytokine interleukin (IL)-37 is an anti-inflammatory member of the IL-1 family of cytokines. Transgenic expression of IL-37 in mice protects them from diet-induced obesity and associated metabolic complications including dyslipidemia, inflammation and insulin resistance. The precise mechanism of action leading to these beneficial metabolic effects is not entirely known. Therefore, we aimed to assess in detail the effect of transgenic IL-37 expression on energy balance, including food intake and energy expenditure. Feeding homozygous IL-37 transgenic mice and wild-type (WT) control mice a high-fat diet (HFD; 45% kcal palm fat) for 6 weeks showed that IL-37 reduced body weight related to a marked decrease in food intake. Subsequent mechanistic studies in mice with heterozygous IL-37 expression versus WT littermates, fed the HFD for 18 weeks, confirmed that IL-37 reduces food intake, which led to a decrease in lean body mass, but did not reduce fat mass and plasma lipid levels or alterations in energy expenditure independent of lean body mass. Taken together, this suggests that IL-37 reduces lean body mass by reducing food intake.
Collapse
|
28
|
de Sousa JR, Prudente RL, Dias Junior LB, Oliveira Carneiro FR, Sotto MN, Simões Quaresma JA. IL-37 and leprosy: A novel cytokine involved in the host response to Mycobacterium leprae infection. Cytokine 2018; 106:89-94. [DOI: 10.1016/j.cyto.2017.10.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/17/2017] [Accepted: 10/18/2017] [Indexed: 12/11/2022]
|
29
|
The Effects of Selective Hematopoietic Expression of Human IL-37 on Systemic Inflammation and Atherosclerosis in LDLr-Deficient Mice. Int J Mol Sci 2017; 18:ijms18081672. [PMID: 28792474 PMCID: PMC5578062 DOI: 10.3390/ijms18081672] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 07/21/2017] [Accepted: 07/28/2017] [Indexed: 12/28/2022] Open
Abstract
The human cytokine interleukin (IL)-37 has potent anti-inflammatory capacities, and hematopoietic cell-specific transgenic overexpression of IL-37 in mice protects against septic shock and colitis. In the present study we investigated the effect of hematopoietic expression of IL-37 on atherosclerosis development under low-grade inflammatory conditions. Low-density lipoprotein receptor (LDLr)-deficient mice were lethally irradiated and transplanted with bone marrow from IL-37-transgenic or control wild-type mice and fed a Western-type diet (WTD; 1% cholesterol) for eight weeks. Metabolic and inflammatory parameters were monitored and atherosclerosis was assessed in the aortic valve area. Hematopoietic IL-37 expression did not influence body weight, food intake and plasma cholesterol levels during the study. Plasma soluble E-selectin levels were increased with WTD-feeding as compared to chow-feeding, but were not influenced by IL-37 expression. IL-37 expression reduced the inflammatory state as indicated by reduced white blood cell counts and by reduced basal and lipopolysaccharide-induced cytokine response by peritoneal macrophages ex vivo. IL-37 expression did not influence the atherosclerotic lesion area. Lesion composition was marginally affected. Smooth muscle cell content was decreased, but macrophage and collagen content were not different. We conclude that under low-grade inflammatory conditions, hematopoietic IL-37 expression reduces the inflammatory state, but does not influence atherosclerosis development in hyperlipidemic LDLr-deficient mice.
Collapse
|
30
|
Zhao M, Hu Y, Jin J, Yu Y, Zhang S, Cao J, Zhai Y, Wei R, Shou J, Cai W, Liu S, Yang X, Xu GT, Yang J, Corry DB, Su SB, Liu X, Yang T. Interleukin 37 promotes angiogenesis through TGF-β signaling. Sci Rep 2017; 7:6113. [PMID: 28733640 PMCID: PMC5522482 DOI: 10.1038/s41598-017-06124-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 05/30/2017] [Indexed: 01/10/2023] Open
Abstract
IL-37 is a novel pro-angiogenic cytokine that potently promotes endothelial cell activation and pathological angiogenesis in our previous study, but the mechanisms behind the pro-angiogenic effect of IL-37 are less well understood. Extending our observations, we found that TGF-β interacts with IL-37, and potently enhances the binding affinity of IL-37 to the ALK1 receptor complex, thus allowing IL-37 to signal through ALK1 to activate pro-angiogenic responses. We further show that TGF-β and ALK1 are required in IL-37 induced pro-angiogenic response in ECs and in the mouse model of Matrigel plug and oxygen-induced retinopathy. The result suggests that IL-37 induces pro-angiogenic responses through TGF-β, which may act as the bridging molecule that mediates IL-37 binding to the TGF-β receptor complex.
Collapse
Affiliation(s)
- Mengmeng Zhao
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yongguang Hu
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiayi Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ying Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shanshan Zhang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingjing Cao
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuanfen Zhai
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rongbin Wei
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Juanjuan Shou
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenping Cai
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shangfeng Liu
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoping Yang
- Johns Hopkins University School of Medicine, Baltimore, United States
| | - Guo-Tong Xu
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianhua Yang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - David B Corry
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States
| | - Shao Bo Su
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xialin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Tianshu Yang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
31
|
Hara T, Monguchi T, Iwamoto N, Akashi M, Mori K, Oshita T, Okano M, Toh R, Irino Y, Shinohara M, Yamashita Y, Shioi G, Furuse M, Ishida T, Hirata KI. Targeted Disruption of JCAD (Junctional Protein Associated With Coronary Artery Disease)/KIAA1462, a Coronary Artery Disease-Associated Gene Product, Inhibits Angiogenic Processes In Vitro and In Vivo. Arterioscler Thromb Vasc Biol 2017; 37:1667-1673. [PMID: 28705794 DOI: 10.1161/atvbaha.117.309721] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 06/30/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Recent genome-wide association studies newly identified the human KIAA1462 gene as a new locus for coronary artery disease. However, the function of the gene product, named JCAD (junctional protein associated with coronary artery disease), is unknown. Because JCAD is expressed at cell-cell junctions in endothelial cells, we hypothesized and tested whether JCAD regulates angiogenic processes in vitro and in vivo. APPROACH AND RESULTS Cell culture experiments revealed impaired angiogenic ability (proliferation, migration, and cord formation) by the knockdown of JCAD with siRNA (P<0.05 versus control siRNA). We have generated mice lacking JCAD (mKIAA1462-/-) by gene-targeted deletion of JCAD to address in vivo angiogenic function. mKIAA1462-/- mice did not show morphological differences in development of retinal vasculature. Ex vivo aortic ring model demonstrated impaired neovascularization in aorta from mKIAA1462-/- mice than control wild-type mice (P<0.05). Tumor growth was assessed by monitoring tumor volume after the subcutaneous injection of melanoma, LLC (Lewis lung carcinoma), and E0771 cells into the mice. mKIAA1462-/- mice exhibited significantly smaller tumor volume compared with wild-type mice (P<0.001). Histological assessment of the tumor exhibited less smooth muscle actin-positive neovascularization determined by CD31-positive vascular structure in tumor of mKIAA1462-/- mice than wild-type mice, indicating that knockdown of JCAD inhibited the vascular maturation in pathological angiogenic process. CONCLUSIONS These in vitro and in vivo studies suggest that JCAD has a redundant functional role in physiological angiogenesis but serves a pivotal role in pathological angiogenic process after birth.
Collapse
MESH Headings
- Animals
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/metabolism
- Cell Adhesion Molecules/deficiency
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Endothelial Cells/metabolism
- Genotype
- Human Umbilical Vein Endothelial Cells/metabolism
- Intercellular Junctions/metabolism
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/genetics
- Melanoma, Experimental/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Neovascularization, Pathologic
- Neovascularization, Physiologic
- Phenotype
- RNA Interference
- Retinal Neovascularization
- Signal Transduction
- Time Factors
- Tissue Culture Techniques
- Transfection
- Tumor Burden
Collapse
Affiliation(s)
- Tetsuya Hara
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.).
| | - Tomoko Monguchi
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| | - Noriko Iwamoto
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| | - Masaya Akashi
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| | - Kenta Mori
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| | - Toshihiko Oshita
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| | - Mitsumasa Okano
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| | - Ryuji Toh
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| | - Yasuhiro Irino
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| | - Masakazu Shinohara
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| | - Yui Yamashita
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| | - Go Shioi
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| | - Mikio Furuse
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| | - Tatsuro Ishida
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| | - Ken-Ichi Hirata
- From the Division of Cardiovascular Medicine, Department of Internal Medicine (T.H., T.M., K.M., T.O., M.O., T.I., K.-i.H.), Division of Cell Biology, Department of Physiology and Cell Biology (N.I., M.A., M.F.), Department of Oral and Maxillofacial Surgery (M.A.), Division of Evidence-Based Laboratory Medicine (R.T., Y.I.), Division of Integrated Medical Education, Department of Community Medicine and Social Healthcare Science (M.S.), and The Integrated Center for Mass Spectrometry (M.S.), Kobe University Graduate School of Medicine, Japan; Animal Resource Development Unit (Y.Y.) and Genetic Engineering Team (Y.Y., G.S.), RIKEN Center for Life Science Technologies, Kobe, Hyogo, Japan; and Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan (M.F.)
| |
Collapse
|
32
|
Zhuang X, Wu B, Li J, Shi H, Jin B, Luo X. The emerging role of interleukin-37 in cardiovascular diseases. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:373-379. [PMID: 28548248 PMCID: PMC5569376 DOI: 10.1002/iid3.159] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 02/09/2017] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Interleukin (IL)-37 is a newly identified member of the IL-1 family, and shows a growing role in a variety of diseases. This review aims at summarizing and discussing the role of IL-37 in cardiovascular diseases. METHODS Data for this review were identified by searches of MEDLINE, Embase, and PubMed using appropriate search terms. RESULTS IL-37 is a newly identified cytokine belonging to the IL-1 family and is expressed in inflammatory immune cells and several parenchymal cells. It has potent anti-inflammatory and immunosuppressive properties, with two mechanisms underlying this function. IL-37 is produced as a precursor and then cleaved into mature form in the cytoplasm by caspase-1, translocating to nucleus and suppressing the transcription of several pro-inflammatory genes by binding SMAD-3. Besides, IL-37 can be secreted extracellularly, and binds to IL-18Ra chain and recruits Toll/IL-1R (TIR)-8 for transducing anti-inflammatory signaling. IL-37 is upregulated in an inducible manner and negatively regulates signaling mediated by TLR agonists and pro-inflammatory cytokines. The cytokine has been shown to inhibit both innate and adaptive immunological responses, exert antitumor effects, and act as a prognostic marker in a variety of autoimmune diseases. CONCLUSIONS Recent studies have suggested that IL-37 plays a role in cardiovascular diseases. In this review, we provide an overview of the cytokine biology, discuss recent advances made in unraveling its cardio-protective effects, and suggest guidelines for future research.
Collapse
Affiliation(s)
- Xinyu Zhuang
- Department of CardiologyHuashan HospitalFudan UniversityShanghaiChina
| | - Bangwei Wu
- Department of CardiologyHuashan HospitalFudan UniversityShanghaiChina
| | - Jian Li
- Department of CardiologyHuashan HospitalFudan UniversityShanghaiChina
| | - Haiming Shi
- Department of CardiologyHuashan HospitalFudan UniversityShanghaiChina
| | - Bo Jin
- Department of CardiologyHuashan HospitalFudan UniversityShanghaiChina
| | - Xinping Luo
- Department of CardiologyHuashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
33
|
Liu H, Ge B. Interleukin-37: a new molecular target for host-directed therapy of tuberculosis. Future Microbiol 2017; 12:465-468. [PMID: 28481119 DOI: 10.2217/fmb-2017-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Haipeng Liu
- Shanghai TB Key Laboratory, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China.,Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Baoxue Ge
- Shanghai TB Key Laboratory, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China.,Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| |
Collapse
|
34
|
Sharma J, Bhar S, Devi CS. A review on interleukins: The key manipulators in rheumatoid arthritis. Mod Rheumatol 2017; 27:723-746. [DOI: 10.1080/14397595.2016.1266071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jatin Sharma
- School of Biosciences and Technology, VIT University, Vellore, India
| | - Sutonuka Bhar
- School of Biosciences and Technology, VIT University, Vellore, India
| | - C. Subathra Devi
- School of Biosciences and Technology, VIT University, Vellore, India
| |
Collapse
|