1
|
Liu J, Yang K, Lin X, Xu J, Cui X, Hao J, Wang W, Wang W, Li L, Hao M. IL-32/NFκB/miR-205 loop sustains the high expression of IL-32 and enhances the motility of cervical cancer cells. Hum Cell 2024; 37:1434-1445. [PMID: 38902566 DOI: 10.1007/s13577-024-01094-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 05/29/2024] [Indexed: 06/22/2024]
Abstract
Human papillomavirus (HPV) infection is a major contributor to cervical cancer. Persistent HPV infection can trigger the expression of IL-32, yet the precise role of IL-32 in the occurrence and development of cervical cancer remains elusive. To investigate this, qRT‒PCR and western blotting were utilized to measure the mRNA and protein expression levels; bioinformatics analysis was used to screen differentially expressed miRNAs; wound healing and transwell assays were conducted to evaluate cell migration and invasion capabilities. Comparative analysis revealed significantly elevated IL-32 expression in cervical cancer tissues and cell lines compared to control groups. In SiHa and/or HeLa, overexpression of IL-32 and IL-32 exposure markedly upregulated miR-205, whereas its knockdown resulted in a substantial downregulation of miR-205. Furthermore, miR-205 also could significantly regulate the expression of IL-32 in HeLa and SiHa cells. Upregulation and downregulation of IL-32 led to a significant increase or decrease in NFκB expression, respectively. Treatment with BAY11-7082 (an NFκB inhibitor) notably decreased miR-205 expression but had no effect on IL-32 levels. qRT‒PCR and western blotting analyses demonstrated that both overexpression and underexpression of IL-32 and miR-205 significantly enhanced or reduced MMP2 and MMP9 expression in cervical cancer cells, respectively. Knockdown of IL-32 significantly inhibited the migration and invasion of HeLa and SiHa; conversely, treatment with rIL-32α and rIL-32γ notably promoted their migration and invasion. In brief, IL-32 is highly expressed via the formation of a positive regulatory loop with NFκB/miR-205, contributing to the persistence of inflammation and promoting the progression of cervical cancer.
Collapse
Affiliation(s)
- Jianbing Liu
- Departments of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, 036000, Shanxi, China
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Kai Yang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiaoyu Lin
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jing Xu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiaohua Cui
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jianqing Hao
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Wei Wang
- Departments of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, 036000, Shanxi, China
| | - Wenhao Wang
- Departments of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, 036000, Shanxi, China
| | - Li Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Min Hao
- Departments of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, Taiyuan, 036000, Shanxi, China.
| |
Collapse
|
2
|
Sathe A, Ayala C, Bai X, Grimes SM, Lee B, Kin C, Shelton A, Poultsides G, Ji HP. GITR and TIGIT immunotherapy provokes divergent multicellular responses in the tumor microenvironment of gastrointestinal cancers. Genome Med 2023; 15:100. [PMID: 38008725 PMCID: PMC10680277 DOI: 10.1186/s13073-023-01259-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 11/14/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Understanding the mechanistic effects of novel immunotherapy agents is critical to improving their successful clinical translation. These effects need to be studied in preclinical models that maintain the heterogenous tumor microenvironment (TME) and dysfunctional cell states found in a patient's tumor. We investigated immunotherapy perturbations targeting co-stimulatory molecule GITR and co-inhibitory immune checkpoint TIGIT in a patient-derived ex vivo system that maintains the TME in its near-native state. Leveraging single-cell genomics, we identified cell type-specific transcriptional reprogramming in response to immunotherapy perturbations. METHODS We generated ex vivo tumor slice cultures from fresh surgical resections of gastric and colon cancer and treated them with GITR agonist or TIGIT antagonist antibodies. We applied paired single-cell RNA and TCR sequencing to the original surgical resections, control, and treated ex vivo tumor slice cultures. We additionally confirmed target expression using multiplex immunofluorescence and validated our findings with RNA in situ hybridization. RESULTS We confirmed that tumor slice cultures maintained the cell types, transcriptional cell states and proportions of the original surgical resection. The GITR agonist was limited to increasing effector gene expression only in cytotoxic CD8 T cells. Dysfunctional exhausted CD8 T cells did not respond to GITR agonist. In contrast, the TIGIT antagonist increased TCR signaling and activated both cytotoxic and dysfunctional CD8 T cells. This included cells corresponding to TCR clonotypes with features indicative of potential tumor antigen reactivity. The TIGIT antagonist also activated T follicular helper-like cells and dendritic cells, and reduced markers of immunosuppression in regulatory T cells. CONCLUSIONS We identified novel cellular mechanisms of action of GITR and TIGIT immunotherapy in the patients' TME. Unlike the GITR agonist that generated a limited transcriptional response, TIGIT antagonist orchestrated a multicellular response involving CD8 T cells, T follicular helper-like cells, dendritic cells, and regulatory T cells. Our experimental strategy combining single-cell genomics with preclinical models can successfully identify mechanisms of action of novel immunotherapy agents. Understanding the cellular and transcriptional mechanisms of response or resistance will aid in prioritization of targets and their clinical translation.
Collapse
Affiliation(s)
- Anuja Sathe
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, CCSR 2245, 269 Campus Drive, Stanford, CA, 94305, USA
| | - Carlos Ayala
- Division of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Xiangqi Bai
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, CCSR 2245, 269 Campus Drive, Stanford, CA, 94305, USA
| | - Susan M Grimes
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, CCSR 2245, 269 Campus Drive, Stanford, CA, 94305, USA
| | - Byrne Lee
- Division of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Cindy Kin
- Division of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Andrew Shelton
- Division of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA, USA
| | - George Poultsides
- Division of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, CCSR 2245, 269 Campus Drive, Stanford, CA, 94305, USA.
| |
Collapse
|
3
|
Hough JT, Zhao L, Lequio M, Heslin AJ, Xiao H, Lewis CC, Zhang J, Bai Q, Wakefield MR, Fang Y. IL-32 and its Paradoxical Role in Neoplasia. Crit Rev Oncol Hematol 2023; 186:104011. [PMID: 37105370 DOI: 10.1016/j.critrevonc.2023.104011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/01/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023] Open
Abstract
Interleukin-32 (IL-32) is an interleukin cytokine usually linked to inflammation. In recent years, it has been found that IL-32 exhibits both pro- and anti-tumor effects. Although most of those effects from IL-32 appear to favor tumor growth, some isoforms have shown to favor tumor suppression. This suggests that the role of IL-32 in neoplasia is very complex. Thus, the role of IL-32 in these various cancers and protein pathways makes it a very crucial component to consider when looking at potential therapeutic options in tumor treatment. In this review, we will explore what is currently known about IL-32, including its relationship with tumorigenesis and the potential for IL-32 to enhance local and systemic anti-tumor immune responses. Such a study might be helpful to accelerate the development of IL-32-based immunotherapies.
Collapse
Affiliation(s)
- Jacob T Hough
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA, 50312; Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212
| | - Lei Zhao
- The Department of Respiratory Medicine, the 2nd People's Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Marco Lequio
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212
| | - Aidan J Heslin
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212
| | - Huaping Xiao
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA, 50312; Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55902
| | - Cade C Lewis
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212
| | - Justin Zhang
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212
| | - Qian Bai
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212
| | - Mark R Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University, Des Moines, IA, 50312; Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212.
| |
Collapse
|
4
|
Sathe A, Ayala C, Bai X, Grimes SM, Lee B, Kin C, Shelton A, Poultsides G, Ji HP. GITR and TIGIT immunotherapy provokes divergent multi-cellular responses in the tumor microenvironment of gastrointestinal cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.13.532299. [PMID: 36993756 PMCID: PMC10054933 DOI: 10.1101/2023.03.13.532299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Understanding the cellular mechanisms of novel immunotherapy agents in the human tumor microenvironment (TME) is critical to their clinical success. We examined GITR and TIGIT immunotherapy in gastric and colon cancer patients using ex vivo slice tumor slice cultures derived from cancer surgical resections. This primary culture system maintains the original TME in a near-native state. We applied paired single-cell RNA and TCR sequencing to identify cell type specific transcriptional reprogramming. The GITR agonist was limited to increasing effector gene expression only in cytotoxic CD8 T cells. The TIGIT antagonist increased TCR signaling and activated both cytotoxic and dysfunctional CD8 T cells, including clonotypes indicative of potential tumor antigen reactivity. The TIGIT antagonist also activated T follicular helper-like cells and dendritic cells, and reduced markers of immunosuppression in regulatory T cells. Overall, we identified cellular mechanisms of action of these two immunotherapy targets in the patients' TME.
Collapse
Affiliation(s)
- Anuja Sathe
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Carlos Ayala
- Division of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA, United States
| | - Xiangqi Bai
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Susan M. Grimes
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Byrne Lee
- Division of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA, United States
| | - Cindy Kin
- Division of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA, United States
| | - Andrew Shelton
- Division of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA, United States
| | - George Poultsides
- Division of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA, United States
| | - Hanlee P. Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
5
|
Ribonucleic Acid Engineering of Dendritic Cells for Therapeutic Vaccination: Ready 'N Able to Improve Clinical Outcome? Cancers (Basel) 2020; 12:cancers12020299. [PMID: 32012714 PMCID: PMC7072269 DOI: 10.3390/cancers12020299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/06/2020] [Accepted: 01/19/2020] [Indexed: 02/06/2023] Open
Abstract
Targeting and exploiting the immune system has become a valid alternative to conventional options for treating cancer and infectious disease. Dendritic cells (DCs) take a central place given their role as key orchestrators of immunity. Therapeutic vaccination with autologous DCs aims to stimulate the patient's own immune system to specifically target his/her disease and has proven to be an effective form of immunotherapy with very little toxicity. A great amount of research in this field has concentrated on engineering these DCs through ribonucleic acid (RNA) to improve vaccine efficacy and thereby the historically low response rates. We reviewed in depth the 52 clinical trials that have been published on RNA-engineered DC vaccination, spanning from 2001 to date and reporting on 696 different vaccinated patients. While ambiguity prevents reliable quantification of effects, these trials do provide evidence that RNA-modified DC vaccination can induce objective clinical responses and survival benefit in cancer patients through stimulation of anti-cancer immunity, without significant toxicity. Succinct background knowledge of RNA engineering strategies and concise conclusions from available clinical and recent preclinical evidence will help guide future research in the larger domain of DC immunotherapy.
Collapse
|
6
|
Abstract
Interleukin-32 (IL-32) was originally identified in natural killer (NK) cells activated by IL-2 in 1992. Thus, it was named NK cell transcript 4 (NK4) because of its unknown function at that time. The function of IL-32 has been elucidated over the last decade. IL-32 is primarily considered to be a booster of inflammatory reactions because it is induced by pro-inflammatory cytokines and stimulates the production of those cytokines and vice versa. Therefore, many studies have been devoted to studying the roles of IL-32 in inflammation-associated cancers, including gastric, colon cancer, and hepatocellular carcinoma. At the same time, roles of IL-32 have also been discovered in other cancers. Collectively, IL-32 fosters the tumor progression by nuclear factor-κB (NF-κB)-mediated cytokines and metalloproteinase production, as well as stimulation of differentiation into immunosuppressive cell types in some cancer types. However, it is also able to induce tumor cell apoptosis and enhance NK and cytotoxic T cell sensitivity in other cancer types. In this review, we will address the function of each IL-32 isoform in different cancer types studied to date, and suggest further strategies to comprehensively elucidate the roles of IL-32 in a context-dependent manner.
Collapse
Affiliation(s)
- Sora Han
- Research Institute for Women's Health, Sookmyung Women's University, Seoul 04310, Korea
| | - Young Yang
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| |
Collapse
|
7
|
Paz H, Tsoi J, Kalbasi A, Grasso CS, McBride WH, Schaue D, Butterfield LH, Maurer DM, Ribas A, Graeber TG, Economou JS. Interleukin 32 expression in human melanoma. J Transl Med 2019; 17:113. [PMID: 30953519 PMCID: PMC6449995 DOI: 10.1186/s12967-019-1862-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 03/27/2019] [Indexed: 02/06/2023] Open
Abstract
Background Various proinflammatory cytokines can be detected within the melanoma tumor microenvironment. Interleukin 32 (IL32) is produced by T cells, NK cells and monocytes/macrophages, but also by a subset of melanoma cells. We sought to better understand the biology of IL32 in human melanoma. Methods We analyzed RNA sequencing data from 53 in-house established human melanoma cell lines and 479 melanoma tumors from The Cancer Genome Atlas dataset. We evaluated global gene expression patterns associated with IL32 expression. We also evaluated the impact of proinflammatory molecules TNFα and IFNγ on IL32 expression and dedifferentiation in melanoma cell lines in vitro. In order to study the transcriptional regulation of IL32 in these cell lines, we cloned up to 10.5 kb of the 5′ upstream region of the human IL32 gene into a luciferase reporter vector. Results A significant proportion of established human melanoma cell lines express IL32, with its expression being highly correlated with a dedifferentiation genetic signature (high AXL/low MITF). Non IL32-expressing differentiated melanoma cell lines exposed to TNFα or IFNγ can be induced to express the three predominant isoforms (α, β, γ) of IL32. Cis-acting elements within this 5′ upstream region of the human IL32 gene appear to govern both induced and constitutive gene expression. In the tumor microenvironment, IL32 expression is highly correlated with genes related to T cell infiltration, and also positively correlates with high AXL/low MITF dedifferentiated gene signature. Conclusions Expression of IL32 in human melanoma can be induced by TNFα or IFNγ and correlates with a treatment-resistant dedifferentiated genetic signature. Constitutive and induced expression are regulated, in part, by cis-acting sequences within the 5′ upstream region. Electronic supplementary material The online version of this article (10.1186/s12967-019-1862-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Helicia Paz
- Department of Surgery, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA
| | - Jennifer Tsoi
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA, 90095, USA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Anusha Kalbasi
- Department of Surgery, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA.,Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA
| | - Catherine S Grasso
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - William H McBride
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Lisa H Butterfield
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA.,Department of Surgery, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA.,Department of Clinical and Translational Science, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Deena M Maurer
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Antoni Ribas
- Department of Surgery, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA, 90095, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA
| | - James S Economou
- Department of Surgery, University of California, Los Angeles, 10833 Le Conte Ave, Los Angeles, CA, 90095, USA. .,Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA. .,Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA, 90095, USA. .,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
8
|
Yan H, He D, Huang X, Zhang E, Chen Q, Xu R, Liu X, Zi F, Cai Z. Role of interleukin-32 in cancer biology. Oncol Lett 2018; 16:41-47. [PMID: 29930712 DOI: 10.3892/ol.2018.8649] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/30/2018] [Indexed: 12/13/2022] Open
Abstract
Interleukin-32 (IL-32), a novel proinflammatory cytokine, is highly expressed in various cancer tissues and in established cancer cell lines. IL-32 has been revealed to serve a crucial role in human cancer development, including tumour initiation, proliferation and maintenance. The expression of IL-32 is regulated by numerous factors, including genetic variations, hypoxia and acidosis in the tumour microenvironment. Understanding the underlying mechanisms of IL-32 expression and its function are critical for the discovery of novel therapeutic strategies that target IL-32. This is a review of the current literature on the regulation and function of IL-32 in cancer progression, focusing on the molecular pathways linking IL-32 and tumour development.
Collapse
Affiliation(s)
- Haimeng Yan
- Bone Marrow Transplantation Centre, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Donghua He
- Bone Marrow Transplantation Centre, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xi Huang
- Bone Marrow Transplantation Centre, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Enfan Zhang
- Bone Marrow Transplantation Centre, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Qingxiao Chen
- Bone Marrow Transplantation Centre, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Ruyi Xu
- Bone Marrow Transplantation Centre, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xinling Liu
- Bone Marrow Transplantation Centre, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Fuming Zi
- Department of Haematology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| | - Zhen Cai
- Bone Marrow Transplantation Centre, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
9
|
Gonnet J, Perrin H, Hutton AJ, Boccara D, Bonduelle O, Mimoun M, Atlan M, Soria A, Combadière B. Interleukin-32 promotes detachment and activation of human Langerhans cells in a human skin explant model. Br J Dermatol 2018; 179:145-153. [PMID: 29806155 DOI: 10.1111/bjd.16721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Cross-talk between skin keratinocytes (KCs) and Langerhans cells (LCs) plays a fundamental role in the body's first line of immunological defences. However, the mechanism behind the interaction between these two major epidermal cells is unknown. Interleukin (IL)-32 is produced in inflammatory skin disorders. We questioned the role of IL-32 in the epidermis. OBJECTIVES We aimed to determine the role of IL-32 produced by KCs on surrounding LCs. METHODS We used an ex vivo human explant model from healthy donors and investigated the role of IL-32 on LC activation using imaging, flow cytometry, reverse transcriptase quantitative polymerase chain reaction and small interfering (si)RNA treatment. RESULTS Modified vaccinia virus ankara (MVA) infection induced KC death alongside the early production of the proinflammatory cytokine IL-32. We demonstrated that IL-32 produced by MVA-infected KCs induced modest but significant morphological changes in LCs and downregulation of adhesion molecules, such as epithelial cell adhesion molecule and very late antigen-4, and CXCL10 production. The treatment of KCs with IL-32-specific siRNA, and anti-IL-32 blocking antibody significantly inhibited LC activation, demonstrating the role of IL-32 in LC activation. We also found that some Toll-like receptor ligands induced a very high level of IL-32 production by KCs, which initiated LC activation. CONCLUSIONS We propose, for the first time, that IL-32 is a molecular link between KCs and LCs in healthy skin, provoking LC migration from the epidermis to the dermis prior to their migration to the draining lymph nodes.
Collapse
Affiliation(s)
- J Gonnet
- Sorbonne Universités UPMC Université Paris 06, UMRS CR7, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses-Paris (Cimi-Paris), 91 Boulevard de l'Hôpital, 75013, Paris, France
| | - H Perrin
- Sorbonne Universités UPMC Université Paris 06, UMRS CR7, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses-Paris (Cimi-Paris), 91 Boulevard de l'Hôpital, 75013, Paris, France
| | - A J Hutton
- Sorbonne Universités UPMC Université Paris 06, UMRS CR7, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses-Paris (Cimi-Paris), 91 Boulevard de l'Hôpital, 75013, Paris, France
| | - D Boccara
- Sorbonne Universités UPMC Université Paris 06, UMRS CR7, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses-Paris (Cimi-Paris), 91 Boulevard de l'Hôpital, 75013, Paris, France.,Service de Chirurgie Plastique Reconstructrice, Esthétique, Centre des Brûlés, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP), 1 avenue Claude Vellefaux, 75010, Paris, France
| | - O Bonduelle
- Sorbonne Universités UPMC Université Paris 06, UMRS CR7, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses-Paris (Cimi-Paris), 91 Boulevard de l'Hôpital, 75013, Paris, France
| | - M Mimoun
- Service de Chirurgie Plastique Reconstructrice, Esthétique, Centre des Brûlés, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP), 1 avenue Claude Vellefaux, 75010, Paris, France
| | - M Atlan
- Service de Chirurgie Plastique Reconstructrice et Esthétique, Hôpital Tenon, Assistance Publique Hôpitaux de Paris (APHP), 4 Rue de la Chine, 75020, Paris, France
| | - A Soria
- Sorbonne Universités UPMC Université Paris 06, UMRS CR7, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses-Paris (Cimi-Paris), 91 Boulevard de l'Hôpital, 75013, Paris, France.,Service de Dermatologie et d'Allergologie, Hôpital Tenon, Hôpitaux Universitaire Est Parisien (HUEP), Assistance Publique Hôpitaux de Paris (APHP), 4 rue de la Chine, 75020, Paris, France
| | - B Combadière
- Sorbonne Universités UPMC Université Paris 06, UMRS CR7, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses-Paris (Cimi-Paris), 91 Boulevard de l'Hôpital, 75013, Paris, France
| |
Collapse
|
10
|
Abstract
Interleukin 32 (IL-32) is a proinflammatory cytokine involved in the development of several diseases, including cancer. IL-32 is a rather peculiar cytokine because its protein structure does not show resemblance with any of the known cytokines, and an IL-32 receptor to facilitate extracellular signaling has not yet been identified. Thus far, 9 isoforms of IL-32 have been described, all of which show differences in terms of effects and in potency to elicit a specific effect. Since the first report of IL-32 in 2005, there is increasing evidence that IL-32 plays an important role in the pathophysiology of both hematologic malignancies and solid tumors. Some IL-32 isoforms have been linked to disease outcome and were shown to positively influence tumor development and progression in various different malignancies, including gastric, breast and lung cancers. However, there are other reports suggesting a tumor suppressive role for some of IL-32 as well. For example, IL-32γ and IL-32β expression is associated with increased cancer cell death in colon cancer and melanoma, whereas expression of these isoforms is associated with increased invasion and migration in breast cancer cells. Furthermore, IL-32 isoforms α, β and γ also play an important role in regulating the anti-tumor immune response, thus also influencing tumor progression. In this review, we provide an overview of the role of IL-32 and its different isoforms in carcinogenesis, invasion and metastasis, angiogenesis and regulation of the anti-tumor immune response.
Collapse
|
11
|
Heinhuis B, Plantinga TS, Semango G, Küsters B, Netea MG, Dinarello CA, Smit JW, Netea-Maier RT, Joosten LA. Alternatively spliced isoforms of IL-32 differentially influence cell death pathways in cancer cell lines. Carcinogenesis 2015; 37:197-205. [DOI: 10.1093/carcin/bgv172] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 12/02/2015] [Indexed: 12/14/2022] Open
|
12
|
IL-32θ inhibits monocytic differentiation of leukemia cells by attenuating expression of transcription factor PU.1. Oncotarget 2015; 6:4394-405. [PMID: 25726525 PMCID: PMC4414198 DOI: 10.18632/oncotarget.3013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 12/31/2014] [Indexed: 12/18/2022] Open
Abstract
PU.1 is a key transcription factor regulating the myeloid differentiation. PU.1-induced monocytic differentiation into macrophage is also important for blood cancer development. Therefore, we chose THP-1 monocytic leukemia cells to investigate the function of a recently discovered IL-32θ. Genetic analyses identified differences in the sequences of IL-32θ and IL-32β. Using previously established cell lines that stably express IL-32θ and IL-32β and cell lines transiently expressing IL-32θ, we observed that expression of IL-32θ inhibited phorbol 12-myristate 13-acetate (PMA)-induced monocytic differentiation in both THP-1 and HL-60 cells. IL-32θ also suppressed expression of the macrophage cell surface markers, CD11b, CD18, and CD36. Interestingly, expression of IL-32β or IL-32θ had no effect on the expression levels of cell cycle related factors. As a result, we concluded that these isoforms did not contribute to PMA-induced cell cycle arrest. IL-32θ was found to modulate expression of PU.1, a transcription factor necessary for myeloid lineage commitment. Transient expression of PU.1 in THP-1/IL-32θ cells rescued the observed differentiation defect. Additionally, transient expression of both CCAAT-enhancer-binding protein α (C/EBPα) and PU.1 in THP-1/IL-32θ cells exhibited synergistic effects in rescuing the differentiation defect. These observations indicate that intracellular IL-32θ inhibits the differentiation of monocytes into macrophages by attenuating PU.1 expression.
Collapse
|
13
|
The Role of IL-32 in Cutaneous T-Cell Lymphoma. J Invest Dermatol 2014; 134:1428-1435. [DOI: 10.1038/jid.2013.488] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 09/30/2013] [Accepted: 10/25/2013] [Indexed: 12/20/2022]
|
14
|
Bhanumathy KK, Zhang B, Ahmed KA, Qureshi M, Xie Y, Tao M, Tan X, Xiang J. Transgene IL-6 enhances DC-stimulated CTL responses by counteracting CD4+25+Foxp3+ regulatory T cell suppression via IL-6-induced Foxp3 downregulation. Int J Mol Sci 2014; 15:5508-21. [PMID: 24690994 PMCID: PMC4013578 DOI: 10.3390/ijms15045508] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 02/25/2014] [Accepted: 03/06/2014] [Indexed: 11/17/2022] Open
Abstract
Dendritic cells (DCs), the most potent antigen-presenting cells have been extensively applied in clinical trials for evaluation of antitumor immunity. However, the efficacy of DC-mediated cancer vaccines is still limited as they are unable to sufficiently break the immune tolerance. In this study, we constructed a recombinant adenoviral vector (AdVIL-6) expressing IL-6, and generated IL-6 transgene-engineered DC vaccine (DCOVA/IL-6) by transfection of murine bone marrow-derived ovalbumin (OVA)-pulsed DCs (DCOVA) with AdVIL-6. We then assessed DCOVA/IL-6-stimulated cytotoxic T-lymphocyte (CTL) responses and antitumor immunity in OVA-specific animal tumor model. We demonstrate that DCOVA/IL-6 vaccine up-regulates expression of DC maturation markers, secretes transgene-encoded IL-6, and more efficiently stimulates OVA-specific CTL responses and therapeutic immunity against OVA-expressing B16 melanoma BL6-10OVA in vivo than the control DCOVA/Null vaccine. Moreover, DCOVA/IL-6-stimulated CTL responses were relatively maintained in mice with transfer of CD4+25+Foxp3+ Tr-cells, but significantly reduced when treated with anti-IL-6 antibody. In addition, we demonstrate that IL-6 down-regulates Foxp3-expression of CD4+25+Foxp3+ Tr-cells in vitro. Taken together, our results demonstrate that AdV-mediated IL-6 transgene-engineered DC vaccine stimulates potent CTL responses and antitumor immunity by counteracting CD4+25+ Tr immunosuppression via IL-6-induced Foxp3 down-regulation. Thus, IL-6 may be a good candidate for engineering DCs for cancer immunotherapy.
Collapse
Affiliation(s)
| | - Bei Zhang
- Cancer Research Unit, Saskatchewan Cancer Agency, Saskatoon, SK S7N 5E5, Canada.
| | | | - Mabood Qureshi
- Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Yufeng Xie
- Department of Oncology, the First Affiliated Hospital of Soochow University, Soochow 215000, China.
| | - Min Tao
- Department of Oncology, the First Affiliated Hospital of Soochow University, Soochow 215000, China.
| | - Xin Tan
- School of Life Sciences, Beijing Institute of Technology, Beijing 100081, China.
| | - Jim Xiang
- Cancer Research Unit, Saskatchewan Cancer Agency, Saskatoon, SK S7N 5E5, Canada.
| |
Collapse
|
15
|
Joosten LAB, Heinhuis B, Netea MG, Dinarello CA. Novel insights into the biology of interleukin-32. Cell Mol Life Sci 2013; 70:3883-92. [PMID: 23463238 PMCID: PMC11113358 DOI: 10.1007/s00018-013-1301-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 02/11/2013] [Accepted: 02/14/2013] [Indexed: 01/25/2023]
Abstract
Interleukin (IL)-32 is known as a proinflammatory cytokine that is likely involved in several diseases, including infections, chronic inflammation, and cancer. Since the first report in 2005, IL-32 has been the subject of numerous studies to unravel the biological function of this molecule. For example, silencing of endogenous IL-32 in primary or cell lines of human origin consistently suppressed responses to Toll-like receptors. The protein folding structure of the six isoforms of IL-32 does not resemble that of any classical cytokine and as of this writing, a specific IL-32 receptor has not been identified. Instead, we propose a mechanism by which exposure to extracellular IL-32 or overexpression of the molecule results in binding to intracellular partners that influences functions such as gene expression, cell death, or survival. As such, this review offers insights into the role of IL-32 in several diseases, host defense, inflammation, immune function, and cancer. Finally, possibilities to target IL-32 in several diseases are proposed.
Collapse
Affiliation(s)
- Leo A B Joosten
- Department of Medicine (463), Radboud University Nijmegen Medical Centre, Geert Grooteplein zuid 8, 6525 GA, Nijmegen, The Netherlands,
| | | | | | | |
Collapse
|
16
|
Abstract
The last decade has seen the emergence of immunomodulators as therapeutic agents in cancer treatment. Interleukins (ILs) are a category of small cell-signaling molecules that organize communication and interaction between immune cells and therefore they could be used as perfect immunomodulators. IL-12 is a promising candidate for cancer immunotherapy since it plays a major role in development of antitumor immune response. Numerous studies report that IL-12 promotes an effective destruction of cancer cells both in vivo and in vitro. In addition, IL-12 has anti-angiogenic activity and it is able to dramatically decrease tumor-supportive activities of tumor-associated macrophages. The first part of the review is devoted to immunobiology of IL-12. Signaling pathways of IL-12 as well as clinical trials of this cytokine are discussed. The second part of the review is concerned on the inherited variations in IL-12A and IL-12B genes that could modulate cancer susceptibility, and as a consequence, possess predictive, therapeutic, or prognostic significance. It is known that functional single nucleotide polymorphisms (SNPs) in IL-12A and IL-12B genes may dramatically affect on protein expression level, or alter its functions, which may lead to immune disorders, autoimmune diseases, and eventually contribute to cancer occurrence. The list of genetic polymorphisms for further investigations might include the following: IL-12B_+1188A/C (rs3212227), IL-12A_+277G/A (rs568408), IL-12A_-798T/A (rs582054), IL-12A_-504T/G (rs190533), IL-12A_-1148T/C (rs2243123), and IL-12B_+16974 A/C. Perhaps, some of these SNPs may become an attractive target for oncogenomics and possibly could be used in programs of early cancer diagnosis as well as cancer prevention in the nearest future.
Collapse
Affiliation(s)
- Arseniy E Yuzhalin
- Department of Genetics, Kemerovo State University, Kemerovo, Russian Federation.
| | | |
Collapse
|
17
|
Therapeutic effect of intratumoral administration of DCs with conditional expression of combination of different cytokines. Cancer Immunol Immunother 2012; 61:573-9. [PMID: 22223258 DOI: 10.1007/s00262-011-1198-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 12/23/2011] [Indexed: 12/14/2022]
Abstract
In this study, we tested the effect of intratumoral administration of dendritic cells (DCs) with inducible expression of different cytokines, using the novel Rheoswitch Therapeutic System on the experimental models of renal cell cancer (RENCA) and MethA sarcoma. Intratumoral injection of DCs, engineered to express IL-12, IL-21, or IFN-α, showed potent therapeutic effect against established tumor. This effect was associated with the induction of potent tumor antigen-specific CD8+ T-cell responses, as well as the infiltration of tumors with CD4+ and CD8+ T cells but not with the cytotoxic activity of DCs. Combination of i.t. administration of DCs, producing different cytokines, did not enhance the antitumor effect of therapy with single cytokine. These results indicate that RTS can be a potent tool for conditional topical cytokine delivery, in combination with DC administration. However, combination of different cytokines may not necessarily improve the outcome of treatment.
Collapse
|