1
|
Arra A, Vogel K, Han I, Behrendt C, Dunay IR, Häupl T, Feist E, Brunner-Weinzierl MC. PD1 + innate lymphoid cells 3 predict JAK-dependent inflammation in rheumatoid arthritis. J Autoimmun 2025; 154:103424. [PMID: 40300483 DOI: 10.1016/j.jaut.2025.103424] [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: 12/20/2024] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
Innate lymphoid cells (ILCs) play a key role in maintaining immune homeostasis and are linked to inflammation and autoimmunity. This study investigates the role of ILCs in the pathogenesis of rheumatoid arthritis (RA) and their response to two targeted therapies - JAK inhibitors (JAKi), which block critical signaling pathways required for their activation, and TNF inhibitors (TNFi), which target a key inflammatory mediator - offering insights into how these interventions shape ILC-driven inflammation. ILC distribution correlated with RA activity, as indicated by the DAS28 score, and this imbalance was improved significantly within four weeks of JAKi, underscoring its early therapeutic impact on ILC-mediated inflammation. While levels of ILC3-activating cytokines such as IL-1β and IL-23 declined under JAKi therapy, they remained unchanged with TNFi. Although JAKi and TNFi showed similar treatment efficacy, multivariate regression analysis showed that improvement in DAS28 score was strongly associated with increase in CTLA-4+ILC3 and reduction in both PD1+ILC3 frequency and systemic IL12p40/IL-23 levels only with JAKi. Notably, this two ILC3 subtypes determined the DAS28 score after 50 d of JAKi. In contrast, patients showing limited response to JAKi (ΔDAS28 < 1.2) maintained high systemic IL-18 levels, a cytokine that induces signaling independent of the JAK pathway, suggesting a potential resistance mechanism. These findings highlight that monitoring PD1+ILC3s or IL-12p40/IL-23 may serve as an indicator of JAKi responsiveness, while elevated IL-18 may identify patients benefiting from alternative therapies. These results also emphasize the clinical relevance of targeting innate immunity for more personalized, pathway-focused RA therapies.
Collapse
Affiliation(s)
- Aditya Arra
- Department of Experimental Pediatrics, Otto-von-Guericke University, 39120, Magdeburg, Germany; Health Campus GC-I3, Medical Faculty, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Katrin Vogel
- Department of Experimental Pediatrics, Otto-von-Guericke University, 39120, Magdeburg, Germany; Health Campus GC-I3, Medical Faculty, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Irina Han
- Department of Experimental Pediatrics, Otto-von-Guericke University, 39120, Magdeburg, Germany; Health Campus GC-I3, Medical Faculty, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Christine Behrendt
- Department of Experimental Rheumatology, Otto-von-Guericke University, 39120, Magdeburg, Germany; Clinic of Rheumatology, Helios, Vogelsang-Gommern, Germany
| | - Ildiko Rita Dunay
- Health Campus GC-I3, Medical Faculty, Otto-von-Guericke University, 39120, Magdeburg, Germany; Institut of Inflammation and Neurodegeneration, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Thomas Häupl
- Department of Experimental Rheumatology, Otto-von-Guericke University, 39120, Magdeburg, Germany; Clinic of Rheumatology, Helios, Vogelsang-Gommern, Germany
| | - Eugen Feist
- Health Campus GC-I3, Medical Faculty, Otto-von-Guericke University, 39120, Magdeburg, Germany; Department of Experimental Rheumatology, Otto-von-Guericke University, 39120, Magdeburg, Germany; Clinic of Rheumatology, Helios, Vogelsang-Gommern, Germany
| | - Monika C Brunner-Weinzierl
- Department of Experimental Pediatrics, Otto-von-Guericke University, 39120, Magdeburg, Germany; Health Campus GC-I3, Medical Faculty, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| |
Collapse
|
2
|
Hosaka I, Ikegami I, Mikami T, Sato T, Ogawa T, Mukawa K, Tanaka M, Endo K, Akiyama Y, Ohkawa A, Nakazawa J, Shibata T, Nakajima T, Iba Y, Shiiku C, Sumino S, Koshima R, Takano K, Ichimiya S, Kawaharada N, Furuhashi M. Unraveling Novel Subsets of Lymphocytes Involved in Sac Expansion in the Tertiary Lymphoid Structure Within an Abdominal Aortic Aneurysm. J Am Heart Assoc 2025; 14:e040279. [PMID: 39968802 DOI: 10.1161/jaha.124.040279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Chronic inflammation is involved in the development of abdominal aortic aneurysm (AAA). A tertiary lymphoid structure (TLS) within vascular lesions has recently been focused on for its role in modulation of inflammation in local tissues. We aimed to elucidate the relationships between TLS and pathophysiology of AAA. METHODS Abdominal aortic samples obtained from 37 patients with AAA (men/women: 34/3, age: 72.8±9.9 years) and 15 autopsied patients who died from non-aortic events (men/women: 11/4, age: 65.5±9.8 years) were investigated. RESULTS TLSs in AAA lesions were confirmed by focal infiltration of CD3-positive cells surrounding germinal center-like structures containing CD20-positive cells between the tunica adventitia and tunica media layers. The formation of a TLS was significantly more prevalent in AAA patients than in autopsied patients. The number of TLSs in AAA lesions was positively correlated with sac diameter (r=0.357, P=0.035) and the amount of intraluminal thrombosis (r=0.466, P=0.005). T cells and B cells were predominant cellular populations among CD45+ cells in AAA lesions. There was a significantly positive correlation between the proportions of interfollicular T follicular helper (CD3+CD4+CD45RA-CXCR5+PD-1+) cells and double negative B (CD3-CD19+IgD-CD27-) cells, and they were positively correlated with sac diameter, intraluminal thrombosis, and serum lipids. Deposited single-cell RNA-sequencing data for AAA showed that T follicular helper cells and double negative B cells were associated with lipid metabolism, T cell activation/proliferation and inflammation. CONCLUSIONS The formation of a TLS in AAA lesions is associated with sac diameter and intraluminal thrombosis in connection with interfollicular T follicular helper cells and double negative B cells, which may contribute to the pathophysiology of AAA and might be novel therapeutic targets for the development of AAA.
Collapse
Affiliation(s)
- Itaru Hosaka
- Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Ippei Ikegami
- Department of Human Immunology Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine Sapporo Japan
| | - Takuma Mikami
- Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine Sapporo Medical University School of Medicine Sapporo Japan
| | - Toshifumi Ogawa
- Department of Cardiovascular, Renal and Metabolic Medicine Sapporo Medical University School of Medicine Sapporo Japan
| | - Kei Mukawa
- Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine Sapporo Medical University School of Medicine Sapporo Japan
| | - Keisuke Endo
- Department of Cardiovascular, Renal and Metabolic Medicine Sapporo Medical University School of Medicine Sapporo Japan
| | - Yukinori Akiyama
- Department of Neurosurgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Akihito Ohkawa
- Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Junji Nakazawa
- Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Tsuyoshi Shibata
- Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Tomohiro Nakajima
- Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Yutaka Iba
- Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Chikara Shiiku
- Department of Cardiovascular Surgery Obihiro Hospital, National Hospital Organization Obihiro Japan
| | - Satoshi Sumino
- Department of Cardiovascular Surgery Sapporo Teishinkai Hospital Sapporo Japan
| | - Ryuji Koshima
- Department of Cardiovascular Surgery Sapporo Cardiovascular Clinic Sapporo Japan
| | - Kenichi Takano
- Department of Otolaryngology - Head and Neck Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Shingo Ichimiya
- Department of Human Immunology Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine Sapporo Japan
| | - Nobuyoshi Kawaharada
- Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine Sapporo Medical University School of Medicine Sapporo Japan
| |
Collapse
|
3
|
Hoffmann MH, Kirchner H, Krönke G, Riemekasten G, Bonelli M. Inflammatory tissue priming: novel insights and therapeutic opportunities for inflammatory rheumatic diseases. Ann Rheum Dis 2024; 83:1233-1253. [PMID: 38702177 DOI: 10.1136/ard-2023-224092] [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: 03/12/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
Due to optimised treatment strategies and the availability of new therapies during the last decades, formerly devastating chronic inflammatory diseases such as rheumatoid arthritis or systemic sclerosis (SSc) have become less menacing. However, in many patients, even state-of-the-art treatment cannot induce remission. Moreover, the risk for flares strongly increases once anti-inflammatory therapy is tapered or withdrawn, suggesting that underlying pathological processes remain active even in the absence of overt inflammation. It has become evident that tissues have the ability to remember past encounters with pathogens, wounds and other irritants, and to react more strongly and/or persistently to the next occurrence. This priming of the tissue bears a paramount role in defence from microbes, but on the other hand drives inflammatory pathologies (the Dr Jekyll and Mr Hyde aspect of tissue adaptation). Emerging evidence suggests that long-lived tissue-resident cells, such as fibroblasts, macrophages, long-lived plasma cells and tissue-resident memory T cells, determine inflammatory tissue priming in an interplay with infiltrating immune cells of lymphoid and myeloid origin, and with systemically acting factors such as cytokines, extracellular vesicles and antibodies. Here, we review the current state of science on inflammatory tissue priming, focusing on tissue-resident and tissue-occupying cells in arthritis and SSc, and reflect on the most promising treatment options targeting the maladapted tissue response during these diseases.
Collapse
Affiliation(s)
| | - Henriette Kirchner
- Institute for Human Genetics, Epigenetics and Metabolism Lab, University of Lübeck, Lübeck, Germany
| | - Gerhard Krönke
- Department of Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Michael Bonelli
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| |
Collapse
|
4
|
Zhao L, Jin S, Wang S, Zhang Z, Wang X, Chen Z, Wang X, Huang S, Zhang D, Wu H. Tertiary lymphoid structures in diseases: immune mechanisms and therapeutic advances. Signal Transduct Target Ther 2024; 9:225. [PMID: 39198425 PMCID: PMC11358547 DOI: 10.1038/s41392-024-01947-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/02/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024] Open
Abstract
Tertiary lymphoid structures (TLSs) are defined as lymphoid aggregates formed in non-hematopoietic organs under pathological conditions. Similar to secondary lymphoid organs (SLOs), the formation of TLSs relies on the interaction between lymphoid tissue inducer (LTi) cells and lymphoid tissue organizer (LTo) cells, involving multiple cytokines. Heterogeneity is a distinguishing feature of TLSs, which may lead to differences in their functions. Growing evidence suggests that TLSs are associated with various diseases, such as cancers, autoimmune diseases, transplant rejection, chronic inflammation, infection, and even ageing. However, the detailed mechanisms behind these clinical associations are not yet fully understood. The mechanisms by which TLS maturation and localization affect immune function are also unclear. Therefore, it is necessary to enhance the understanding of TLS development and function at the cellular and molecular level, which may allow us to utilize them to improve the immune microenvironment. In this review, we delve into the composition, formation mechanism, associations with diseases, and potential therapeutic applications of TLSs. Furthermore, we discuss the therapeutic implications of TLSs, such as their role as markers of therapeutic response and prognosis. Finally, we summarize various methods for detecting and targeting TLSs. Overall, we provide a comprehensive understanding of TLSs and aim to develop more effective therapeutic strategies.
Collapse
Affiliation(s)
- Lianyu Zhao
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Song Jin
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Shengyao Wang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Zhe Zhang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Xuan Wang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Zhanwei Chen
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Xiaohui Wang
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Shengyun Huang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| | - Dongsheng Zhang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| | - Haiwei Wu
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| |
Collapse
|
5
|
Guo Q, Jin Y, Chen X, Ye X, Shen X, Lin M, Zeng C, Zhou T, Zhang J. NF-κB in biology and targeted therapy: new insights and translational implications. Signal Transduct Target Ther 2024; 9:53. [PMID: 38433280 PMCID: PMC10910037 DOI: 10.1038/s41392-024-01757-9] [Citation(s) in RCA: 424] [Impact Index Per Article: 424.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 03/05/2024] Open
Abstract
NF-κB signaling has been discovered for nearly 40 years. Initially, NF-κB signaling was identified as a pivotal pathway in mediating inflammatory responses. However, with extensive and in-depth investigations, researchers have discovered that its role can be expanded to a variety of signaling mechanisms, biological processes, human diseases, and treatment options. In this review, we first scrutinize the research process of NF-κB signaling, and summarize the composition, activation, and regulatory mechanism of NF-κB signaling. We investigate the interaction of NF-κB signaling with other important pathways, including PI3K/AKT, MAPK, JAK-STAT, TGF-β, Wnt, Notch, Hedgehog, and TLR signaling. The physiological and pathological states of NF-κB signaling, as well as its intricate involvement in inflammation, immune regulation, and tumor microenvironment, are also explicated. Additionally, we illustrate how NF-κB signaling is involved in a variety of human diseases, including cancers, inflammatory and autoimmune diseases, cardiovascular diseases, metabolic diseases, neurological diseases, and COVID-19. Further, we discuss the therapeutic approaches targeting NF-κB signaling, including IKK inhibitors, monoclonal antibodies, proteasome inhibitors, nuclear translocation inhibitors, DNA binding inhibitors, TKIs, non-coding RNAs, immunotherapy, and CAR-T. Finally, we provide an outlook for research in the field of NF-κB signaling. We hope to present a stereoscopic, comprehensive NF-κB signaling that will inform future research and clinical practice.
Collapse
Affiliation(s)
- Qing Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yizi Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyu Chen
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Shanghai Cancer Institute & Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Xiaomin Ye
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Xin Shen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingxi Lin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cheng Zeng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Teng Zhou
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Zou X, Guan C, Gao J, Shi W, Cui Y, Zhong X. Tertiary lymphoid structures in pancreatic cancer: a new target for immunotherapy. Front Immunol 2023; 14:1222719. [PMID: 37529035 PMCID: PMC10388371 DOI: 10.3389/fimmu.2023.1222719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/26/2023] [Indexed: 08/03/2023] Open
Abstract
Pancreatic cancer (PC) is extremely malignant and shows limited response to available immunotherapies due to the hypoxic and immunosuppressive nature of its tumor microenvironment (TME). The aggregation of immune cells (B cells, T cells, dendritic cells, etc.), which is induced in various chronic inflammatory settings such as infection, inflammation, and tumors, is known as the tertiary lymphoid structure (TLS). Several studies have shown that TLSs can be found in both intra- and peritumor tissues of PC. The role of TLSs in peritumor tissues in tumors remains unclear, though intratumoral TLSs are known to play an active role in a variety of tumors, including PC. The formation of intratumoral TLSs in PC is associated with a good prognosis. In addition, TLSs can be used as an indicator to assess the effectiveness of treatment. Targeted induction of TLS formation may become a new avenue of immunotherapy for PC. This review summarizes the formation, characteristics, relevant clinical outcomes, and clinical applications of TLSs in the pancreatic TME. We aim to provide new ideas for future immunotherapy of PC.
Collapse
Affiliation(s)
- Xinlei Zou
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Canghai Guan
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianjun Gao
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wujiang Shi
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunfu Cui
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangyu Zhong
- Department of Hepatopancreatobiary Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
7
|
Rasmussen JC, Aldrich MB, Fife CE, Herbst KL, Sevick‐Muraca EM. Lymphatic function and anatomy in early stages of lipedema. Obesity (Silver Spring) 2022; 30:1391-1400. [PMID: 35707862 PMCID: PMC9542082 DOI: 10.1002/oby.23458] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Lipedema is an inflammatory subcutaneous adipose tissue disease that develops in women and may progress to lipolymphedema, a condition similar to lymphedema, in which lymphatic dysfunction results in irresolvable edema. Because it has been shown that dilated lymphatic vessels, impaired pumping, and dermal backflow are associated with presymptomatic, cancer-acquired lymphedema, this study sought to understand whether these abnormal lymphatic characteristics also characterize early stages of lipedema prior to lipolymphedema development. METHODS In a pilot study of 20 individuals with Stage I or II lipedema who had not progressed to lipolymphedema, lymphatic vessel anatomy and function in upper and lower extremities were assessed by near-infrared fluorescence lymphatic imaging and compared with that of a control population of similar age and BMI. RESULTS These studies showed that, although lower extremity lymphatic vessels were dilated and showed intravascular pooling, the propulsion rates significantly exceeded those of control individuals. Upper extremity lymphatics of individuals with lipedema were unremarkable. In contrast to individuals with lymphedema, individuals with Stage I and II lipedema did not exhibit dermal backflow. CONCLUSIONS These results suggest that, despite the confusion in the diagnoses between lymphedema and lipedema, their etiologies differ, with lipedema associated with lymphatic vessel dilation but not lymphatic dysfunction.
Collapse
Affiliation(s)
- John C. Rasmussen
- Center for Molecular Imaging, Brown Foundation Institute of Molecular Medicine, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Melissa B. Aldrich
- Center for Molecular Imaging, Brown Foundation Institute of Molecular Medicine, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Caroline E. Fife
- Department of GeriatricsBaylor College of MedicineHoustonTexasUSA
- CHI St. Luke's HospitalThe WoodlandsTexasUSA
| | - Karen L. Herbst
- Department of MedicineUniversity of ArizonaTucsonArizonaUSA
- Present address:
Total Lipedema CareBeverly HillsCaliforniaUSA
- Present address:
Total Lipedema CareTucsonArizonaUSA
| | - Eva M. Sevick‐Muraca
- Center for Molecular Imaging, Brown Foundation Institute of Molecular Medicine, McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| |
Collapse
|
8
|
Lymph node formation and B cell homeostasis require IKK-α in distinct endothelial cell-derived compartments. Proc Natl Acad Sci U S A 2021; 118:2100195118. [PMID: 34810256 DOI: 10.1073/pnas.2100195118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2021] [Indexed: 11/18/2022] Open
Abstract
Global inactivation of IκB kinase (IKK)-α results in defective lymph node (LN) formation and B cell maturation, and loss of IKK-α-dependent noncanonical NF-κB signaling in stromal organizer and hematopoietic cells is thought to underlie these distinct defects. We previously demonstrated that this pathway is also activated in vascular endothelial cells (ECs). To determine the physiologic function of EC-intrinsic IKK-α, we crossed IkkαF/F mice with Tie2-cre or Cdh5-cre mice to ablate IKK-α in ECs. Notably, the compound defects of global IKK-α inactivation were recapitulated in IkkαTie2 and IkkαCdh5 mice, as both lacked all LNs and mature follicular and marginal zone B cell numbers were markedly reduced. However, as Tie2-cre and Cdh5-cre are expressed in all ECs, including blood forming hemogenic ECs, IKK-α was also absent in hematopoietic cells (HC). To determine if loss of HC-intrinsic IKK-α affected LN development, we generated IkkαVav mice lacking IKK-α in only the hematopoietic compartment. While mature B cell numbers were significantly reduced in IkkαVav mice, LN formation was intact. As lymphatic vessels also arise during development from blood ECs, we generated IkkαLyve1 mice lacking IKK-α in lymphatic ECs (LECs) to determine if IKK-α in lymphatic vessels impacts LN development. Strikingly, while mature B cell numbers were normal, LNs were completely absent in IkkαLyve1 mice. Thus, our findings reveal that IKK-α in distinct EC-derived compartments is uniquely required to promote B cell homeostasis and LN development, and we establish that LEC-intrinsic IKK-α is absolutely essential for LN formation.
Collapse
|
9
|
Vella G, Guelfi S, Bergers G. High Endothelial Venules: A Vascular Perspective on Tertiary Lymphoid Structures in Cancer. Front Immunol 2021; 12:736670. [PMID: 34484246 PMCID: PMC8416033 DOI: 10.3389/fimmu.2021.736670] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/30/2021] [Indexed: 01/22/2023] Open
Abstract
High endothelial venules (HEVs) are specialized postcapillary venules composed of cuboidal blood endothelial cells that express high levels of sulfated sialomucins to bind L-Selectin/CD62L on lymphocytes, thereby facilitating their transmigration from the blood into the lymph nodes (LN) and other secondary lymphoid organs (SLO). HEVs have also been identified in human and murine tumors in predominantly CD3+T cell-enriched areas with fewer CD20+B-cell aggregates that are reminiscent of tertiary lymphoid-like structures (TLS). While HEV/TLS areas in human tumors are predominantly associated with increased survival, tumoral HEVs (TU-HEV) in mice have shown to foster lymphocyte-enriched immune centers and boost an immune response combined with different immunotherapies. Here, we discuss the current insight into TU-HEV formation, function, and regulation in tumors and elaborate on the functional implication, opportunities, and challenges of TU-HEV formation for cancer immunotherapy.
Collapse
Affiliation(s)
- Gerlanda Vella
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, Vlaams Instituut voor Biotechnologie (VIB)-Center for Cancer Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Sophie Guelfi
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, Vlaams Instituut voor Biotechnologie (VIB)-Center for Cancer Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, Vlaams Instituut voor Biotechnologie (VIB)-Center for Cancer Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Department of Neurological Surgery, UCSF Comprehensive Cancer Center, University of California San Francisco (UCSF), San Francisco, CA, United States
| |
Collapse
|
10
|
Asam S, Nayar S, Gardner D, Barone F. Stromal cells in tertiary lymphoid structures: Architects of autoimmunity. Immunol Rev 2021; 302:184-195. [PMID: 34060101 DOI: 10.1111/imr.12987] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
The molecular mediators present within the inflammatory microenvironment are able, in certain conditions, to favor the initiation of tertiary lymphoid structure (TLS) development. TLS is organized lymphocyte clusters able to support antigen-specific immune response in non-immune organs. Importantly, chronic inflammation does not always result in TLS formation; instead, TLS has been observed to develop specifically in permissive organs, suggesting the presence of tissue-specific cues that are able to imprint the immune responses and form TLS hubs. Fibroblasts are tissue-resident cells that define the anatomy and function of a specific tissue. Fibroblast plasticity and specialization in inflammatory conditions have recently been unraveled in both immune and non-immune organs revealing a critical role for these structural cells in human physiology. Here, we describe the role of fibroblasts in the context of TLS formation and its functional maintenance in the tissue, highlighting their potential role as therapeutic disease targets in TLS-associated diseases.
Collapse
Affiliation(s)
- Saba Asam
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Saba Nayar
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, UK
| | - David Gardner
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Francesca Barone
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
11
|
De Pasquale C, Campana S, Bonaccorsi I, Carrega P, Ferlazzo G. ILC in chronic inflammation, cancer and targeting with biologicals. Mol Aspects Med 2021; 80:100963. [PMID: 33726947 DOI: 10.1016/j.mam.2021.100963] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 11/11/2020] [Accepted: 03/04/2021] [Indexed: 12/14/2022]
Abstract
Since their discovery, Innate Lymphoid Cells (ILC) have emerged as important effector cells, serving multiple roles in maintaining tissue homeostasis and responding to tissue insults. As such, dysregulations of their function and distribution have been observed in a variety of immune-mediated diseases, suggesting a specific role for ILC in the pathophysiology of several disorders including chronic inflammation and cancer. Here, we provide an updated view on ILC biology dissecting their pathological or protective contribution in chronic inflammatory diseases such as multiple sclerosis, inflammatory bowel diseases, psoriasis, rheumatoid arthritis, asthma and COPD, atherosclerosis, also exploring ILC role in tumor surveillance and progression. Throughout the review, we will also highlight how the potential dual role of these cells for protective or pathogenic immunity in many inflammatory diseases makes them interesting targets for the development of novel therapeutic strategies, particularly promising.
Collapse
Affiliation(s)
- Claudia De Pasquale
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy
| | - Stefania Campana
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy
| | - Irene Bonaccorsi
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy; Cell Factory Center and Division of Clinical Pathology, University Hospital Policlinico G.Martino, Messina, Italy
| | - Paolo Carrega
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy
| | - Guido Ferlazzo
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy; Cell Factory Center and Division of Clinical Pathology, University Hospital Policlinico G.Martino, Messina, Italy.
| |
Collapse
|
12
|
Pflug KM, Sitcheran R. Targeting NF-κB-Inducing Kinase (NIK) in Immunity, Inflammation, and Cancer. Int J Mol Sci 2020; 21:E8470. [PMID: 33187137 PMCID: PMC7696043 DOI: 10.3390/ijms21228470] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/03/2020] [Accepted: 11/07/2020] [Indexed: 12/23/2022] Open
Abstract
NF-κB-inducing kinase (NIK), the essential upstream kinase, which regulates activation of the noncanonical NF-κB pathway, has important roles in regulating immunity and inflammation. In addition, NIK is vital for maintaining cellular health through its control of fundamental cellular processes, including differentiation, growth, and cell survival. As such aberrant expression or regulation of NIK is associated with several disease states. For example, loss of NIK leads to severe immune defects, while the overexpression of NIK is observed in inflammatory diseases, metabolic disorders, and the development and progression of cancer. This review discusses recent studies investigating the therapeutic potential of NIK inhibitors in various diseases.
Collapse
Affiliation(s)
- Kathryn M. Pflug
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX 77843, USA;
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX 77002, USA
| | - Raquel Sitcheran
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX 77843, USA;
- Department of Molecular & Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX 77002, USA
| |
Collapse
|
13
|
Antonioli L, Fornai M, Pellegrini C, Masi S, Puxeddu I, Blandizzi C. Ectopic Lymphoid Organs and Immune-Mediated Diseases: Molecular Basis for Pharmacological Approaches. Trends Mol Med 2020; 26:1021-1033. [PMID: 32600794 DOI: 10.1016/j.molmed.2020.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/21/2020] [Accepted: 06/04/2020] [Indexed: 12/15/2022]
Abstract
Chronic inflammation is the result a persistent increase in the expression of several proinflammatory pathways with impaired inflammatory resolution. Ectopic lymphoid organs (ELOs), untypical lymphoid annexes, emerge during chronic inflammation and contribute to the physiopathology of chronic inflammatory disorders. This review discusses the pathophysiological role of ELOs in the progression of immune-mediated inflammatory diseases (IMIDs), including multiple sclerosis (MS), rheumatoid arthritis (RA), inflammatory bowel disease (IBD), atherosclerosis, and Sjögren syndrome (SSj). The molecular pathways underlying the emergence of ELOs are of interest for the development of novel pharmacological approaches for the management of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy.
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | | | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Ilaria Puxeddu
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
14
|
Fang W, Zhang Y, Chen Z. Innate lymphoid cells in inflammatory arthritis. Arthritis Res Ther 2020; 22:25. [PMID: 32051038 PMCID: PMC7017550 DOI: 10.1186/s13075-020-2115-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 02/04/2020] [Indexed: 12/19/2022] Open
Abstract
Aberrant activation and dysregulation of immune system is a common feature of many forms of inflammatory arthritis. Since their identification as a distinctive population of leukocytes, innate lymphoid cells (ILCs) have been considered crucial in maintaining tissue homeostasis and bridges between innate and adaptive immune system. Altered ILCs’ subset distribution and function have been observed in a variety of autoimmune and chronic inflammatory diseases and suggest a subset-specific role of ILCs in the pathogenesis of immune-mediated inflammation. In this review, we focus on the current knowledge of ILC subset and their role in inflammatory arthritis, including rheumatoid arthritis (RA), ankylosing spondylitis (AS), psoriatic arthritis (PsA), enteropathic arthritis, and other seronegative spondyloarthritis. By better understanding the biology and function of ILC subset in different disease settings, new therapeutic interventions can be anticipated by modulating dysregulated ILC responses toward promoting resolution of inflammation.
Collapse
Affiliation(s)
- Weiting Fang
- Department of Rheumatology and Immunology, Anhui Medical University Affiliated Provincial Hospital, Lujiang Str 17, Hefei, 230001, China
| | - Yuanyuan Zhang
- Department of Rheumatology and Immunology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Lujiang Str 17, Hefei, 230001, China
| | - Zhu Chen
- Department of Rheumatology and Immunology, Anhui Medical University Affiliated Provincial Hospital, Lujiang Str 17, Hefei, 230001, China. .,Department of Rheumatology and Immunology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Lujiang Str 17, Hefei, 230001, China.
| |
Collapse
|
15
|
Abstract
The development of rheumatoid arthritis (RA), at least in its autoantibody-positive subset, evolves through a series of events starting well before the appearance of synovitis. The distinction between 'early' and 'established' RA is, therefore, an evolving concept. In routine practice, however, the management of RA still starts with the occurrence of clinically detectable synovitis. As such, the synovial membrane remains a major target for the exploitation of possible stage-specific drivers of the disease. The recognition of a 'window of opportunity', in which treatment is more likely to succeed, raises the hypothesis that there might be a period in which the biological processes of RA are less mature and potentially reversible. The present review aims to provide a general picture of the modifications occurring in RA synovium, analysing the contribution of both infiltrating immune cells and stromal cells. When available, differences between early and established RA will be discussed.
Collapse
|
16
|
Abstract
The adaptive immune response is a 500-million-year-old (the "Big Bang" of Immunology) collective set of rearranged and/or selected receptors capable of recognizing soluble and cell surface molecules or shape (B cells, antibody), endogenous and extracellular peptides presented by Major Histocompatibility (MHC) molecules including Class I and Class II (conventional αβ T cells), lipid in the context of MHC-like molecules of the CD1 family (NKT cells), metabolites and B7 family molecules/butyrophilins with stress factors (γδT cells), and stress ligands and absence of MHC molecules (natural killer, NK cells). What makes tumor immunogenic is the recruitment of initially innate immune cells to sites of stress or tissue damage with release of Damage-Associated Molecular Pattern (DAMP) molecules. Subsequent maintenance of a chronic inflammatory state, representing a balance between mature, normalized blood vessels, innate and adaptive immune cells and the tumor provides a complex tumor microenvironment serving as the backdrop for Darwinian selection, tumor elimination, tumor equilibrium, and ultimately tumor escape. Effective immunotherapies are still limited, given the complexities of this highly evolved and selected tumor microenvironment. Cytokine therapies and Immune Checkpoint Blockade (ICB) enable immune effector function and are largely dependent on the shape and size of the B and T cell repertoires (the "adaptome"), now accessible by Next-Generation Sequencing (NGS) and dimer-avoidance multiplexed PCR. How immune effectors access the tumor (infiltrated, immune sequestered, and immune desserts), egress and are organized within the tumor are of contemporary interest and substantial investigation.
Collapse
|
17
|
Jeucken KCM, Koning JJ, Mebius RE, Tas SW. The Role of Endothelial Cells and TNF-Receptor Superfamily Members in Lymphoid Organogenesis and Function During Health and Inflammation. Front Immunol 2019; 10:2700. [PMID: 31824495 PMCID: PMC6879661 DOI: 10.3389/fimmu.2019.02700] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/04/2019] [Indexed: 01/02/2023] Open
Abstract
Lymph nodes (LNs) are crucial for the orchestration of immune responses. LN reactions depend on interactions between incoming and local immune cells, and stromal cells. To mediate these cellular interactions an organized vascular network within the LN exists. In general, the LN vasculature can be divided into two components: blood vessels, which include the specialized high endothelial venules that recruit lymphocytes from the bloodstream, and lymphatic vessels. Signaling via TNF receptor (R) superfamily (SF) members has been implicated as crucial for the development and function of LNs and the LN vasculature. In recent years the role of cell-specific signaling of TNFRSF members in different endothelial cell (EC) subsets and their roles in development and maintenance of lymphoid organs has been elucidated. Here, we discuss recent insights into EC-specific TNFRSF member signaling and highlight its importance in different EC subsets in LN organogenesis and function during health, and in lymphocyte activation and tertiary lymphoid structure formation during inflammation.
Collapse
Affiliation(s)
- Kim C M Jeucken
- Amsterdam Rheumatology and Immunology Center (ARC), Department of Rheumatology and Clinical Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jasper J Koning
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center (ARC), Department of Rheumatology and Clinical Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
18
|
Sofopoulos M, Fortis SP, Vaxevanis CK, Sotiriadou NN, Arnogiannaki N, Ardavanis A, Vlachodimitropoulos D, Perez SA, Baxevanis CN. The prognostic significance of peritumoral tertiary lymphoid structures in breast cancer. Cancer Immunol Immunother 2019; 68:1733-1745. [PMID: 31598757 DOI: 10.1007/s00262-019-02407-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/29/2019] [Indexed: 12/31/2022]
Abstract
Tumors and their surrounding area represent spatially organized "ecosystems", where tumor cells and the immune contextures of the different compartments are in a dynamic interplay, with potential clinical impact. Here, we aimed to investigate the prognostic significance of peritumoral tertiary lymphoid structures (TLS) either alone or jointly with the intratumoral densities and spatial distribution of CD8 + and CD163 + cells in breast cancer (BCa) patients. TLS were identified peritumorally, within the area distancing up to 5 mm from the infiltrative tumor border, counted and further characterized as adjacent or distal, in formalin-fixed, paraffin-embedded tumor tissue samples from a cohort of 167 patients, with histologically confirmed invasive ductal BCa. TLS and tumor-infiltrating immune cells were determined by H&E and immunohistochemistry. Clinical follow-up was available for 112 of these patients. Patients with peritumoral TLS exhibited worse disease-free survival (DFS) and overall survival (OS) as compared to patients lacking TLS. Moreover, the density of peritumoral TLS was found to be crucial for prognosis, since patients with abundant TLS exhibited the worst DFS and OS. By combining the density of adjacent TLS (aTLS) with our recently published intratumoral signatures based on the differential distribution of CD8 + and CD163 + in the tumor center and invasive margin, we created two improved immune signatures with superior prognostic strength and higher patient population coverage. Our observations strengthen the notion for the fundamental role of the dynamic interplay between the immune cells within the tumor microenvironment (center/invasive margin) and the tumor surrounding area (peritumoral TLS) on the clinical outcome of BCa patients.
Collapse
Affiliation(s)
| | - Sotirios P Fortis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras Avenue, 11522, Athens, Greece
| | - Christoforos K Vaxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras Avenue, 11522, Athens, Greece
| | | | | | | | | | - Sonia A Perez
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras Avenue, 11522, Athens, Greece
| | - Constantin N Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras Avenue, 11522, Athens, Greece.
| |
Collapse
|
19
|
Takaki-Kuwahara A, Arinobu Y, Miyawaki K, Yamada H, Tsuzuki H, Irino K, Ayano M, Kimoto Y, Mitoma H, Akahoshi M, Tsukamoto H, Horiuchi T, Niiro H, Akashi K. CCR6+ group 3 innate lymphoid cells accumulate in inflamed joints in rheumatoid arthritis and produce Th17 cytokines. Arthritis Res Ther 2019; 21:198. [PMID: 31470891 PMCID: PMC6716915 DOI: 10.1186/s13075-019-1984-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/19/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Recent studies show that innate lymphoid cells (ILCs) contribute to the development of chronic inflammation and autoimmune disease. In this study, we assessed the ILC function in the development of rheumatoid arthritis (RA). METHODS In a mouse model of collagen-induced arthritis (CIA), we identified and purified the ILC subsets in peripheral blood (PB), local lymph nodes (LNs), and joints by fluorescence-activated cell sorting and used quantitative PCR to assess the expression levels of representative cytokines. We also correlated the frequencies of each ILC subset in synovial fluid (SF) with clinical parameters in RA patients. RESULTS In the CIA model, the proportion of CCR6+ ILC3s to total ILCs in joints with active inflammation significantly increased relative to non-arthritic joints (median 29.6% vs 16.7%, p = 0.035). CCR6+ ILC3s from mice with arthritis expressed significantly higher levels of IL-17A and IL-22 mRNA than did comparable cells from control mice (p < 0.0001 and p = 0.015). In RA patients, the proportion of CCR6+ ILCs in SF was positively correlated with tender joint counts (TJC) and swollen joint counts (SJC) (ρ=0.689, p = 0.0032 and ρ=0.644, p = 0.0071, respectively). Levels of CC chemokine ligand 20 (CCL20) increased in SF of patients with RA and were significantly correlated with CCR6+ ILC number (ρ=0.697, p = 0.0001). CONCLUSION CCR6+ ILC3s may play some roles in the development of RA through the production of IL-17 and IL-22.
Collapse
Affiliation(s)
- Ayako Takaki-Kuwahara
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Yojiro Arinobu
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Kohta Miyawaki
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hisakata Yamada
- Division of Host Defense, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hirofumi Tsuzuki
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kensuke Irino
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masahiro Ayano
- Department of Stem Cell Biology and Medicine Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasutaka Kimoto
- Department of Internal Medicine, Kyushu University Beppu Hospital, Beppu, Japan
| | - Hiroki Mitoma
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Mitsuteru Akahoshi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroshi Tsukamoto
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takahiko Horiuchi
- Department of Internal Medicine, Kyushu University Beppu Hospital, Beppu, Japan
| | - Hiroaki Niiro
- Faculty of Medical Sciences Medical Education, Kyushu University, Fukuoka, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
20
|
Khan I, Bhardwaj M, Shukla S, Lee H, Oh MH, Bajpai VK, Huh YS, Kang SC. Carvacrol encapsulated nanocarrier/ nanoemulsion abrogates angiogenesis by downregulating COX-2, VEGF and CD31 in vitro and in vivo in a lung adenocarcinoma model. Colloids Surf B Biointerfaces 2019; 181:612-622. [PMID: 31202132 DOI: 10.1016/j.colsurfb.2019.06.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 11/18/2022]
Abstract
Nanoemulsion-based synthesis has been introduced to enhance the bioavailability of natural compounds at target sites for their various biomedical applications. In this study, we synthesized carvacrol nanoemulsion (CN) an oil-in-water (O/W) as a nano-emulsion vehicle system by using ultrasonication emulsification for anti-angiogenesis therapy formulated by combining MCT, lecithin, and polysorbate 80 at the O/W interface called carvacrol encapsulated nanoemulsion (CEN). The diameter of CEN determined by TEM analysis was 105.32 nm. The hydrodynamic droplet size was 101.0 nm with a -39.38-mV zeta potential. The stability of the synthesized CEN was approved till 100 days without any change in diameter size distribution and encapsulation efficiency. We evaluated the role of CEN on angiogenesis in lung adenocarcinoma A549 cells both in vitro and in vivo and observed that it reduced the growth and MMP levels of A549 cells in a dose-dependent manner. Exposure to CEN decreased the activation of MAPK p38 as well as ERK. Moreover, we found that CEN reduced the expression of VEGF and CD31 in A549 cells both in vitro and in vivo. Our in-silico study also indicated the binding of carvacrol to COX-2 and VEGF at the active and allosteric sites of CD31 with low binding energy. Overall, CEN induced anti-angiogenic effects in A549 cells in vitro, in silico, and in vivo, thereby establishing its potential as targeted drug delivery vehicle against angiogenesis.
Collapse
Affiliation(s)
- Imran Khan
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC), Inha University, 100 Inha-ro, Nam-gu, Incheon, 22212, Republic of Korea; Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Monika Bhardwaj
- Laboratory of Biochemistry and cellular Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 42988, Republic of Korea; Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Shruti Shukla
- Department of Energy and Materials Engineering, Dongguk University-Seoul, 30 Pildong-ro 1-gil, Seoul, 04620, Republic of Korea
| | - Hoomin Lee
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC), Inha University, 100 Inha-ro, Nam-gu, Incheon, 22212, Republic of Korea
| | - Mi-Hwa Oh
- Animal Production Research and Development Division, National Institute of Animal Science, Jeonju, 54875, Republic of Korea
| | - Vivek K Bajpai
- Department of Energy and Materials Engineering, Dongguk University-Seoul, 30 Pildong-ro 1-gil, Seoul, 04620, Republic of Korea.
| | - Yun Suk Huh
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC), Inha University, 100 Inha-ro, Nam-gu, Incheon, 22212, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
21
|
Wenink MH, Leijten EFA, Cupedo T, Radstake TRDJ. Review: Innate Lymphoid Cells: Sparking Inflammatory Rheumatic Disease? Arthritis Rheumatol 2019; 69:885-897. [PMID: 28217945 DOI: 10.1002/art.40068] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 02/07/2017] [Indexed: 02/06/2023]
Affiliation(s)
| | | | - Tom Cupedo
- Erasmus University Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
22
|
Kucharzewska P, Maracle CX, Jeucken KCM, van Hamburg JP, Israelsson E, Furber M, Tas SW, Olsson HK. NIK-IKK complex interaction controls NF-κB-dependent inflammatory activation of endothelium in response to LTβR ligation. J Cell Sci 2019; 132:jcs225615. [PMID: 30837284 DOI: 10.1242/jcs.225615] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/14/2019] [Indexed: 12/26/2022] Open
Abstract
NF-κB-inducing kinase (NIK; also known as MAP3K14) is a central regulator of non-canonical NF-κB signaling in response to stimulation of TNF receptor superfamily members, such as the lymphotoxin-β receptor (LTβR), and is implicated in pathological angiogenesis associated with chronic inflammation and cancer. Here, we identify a previously unrecognized role of the LTβR-NIK axis during inflammatory activation of human endothelial cells (ECs). Engagement of LTβR-triggered canonical and non-canonical NF-κB signaling promoted expression of inflammatory mediators and adhesion molecules, and increased immune cell adhesion to ECs. Sustained LTβR-induced inflammatory activation of ECs was NIK dependent, but independent of p100, indicating that the non-canonical arm of NF-κB is not involved. Instead, prolonged activation of canonical NF-κB signaling, through the interaction of NIK with IκB kinase α and β (also known as CHUK and IKBKB, respectively), was required for the inflammatory response. Endothelial inflammatory activation induced by synovial fluid from rheumatoid arthritis patients was significantly reduced by NIK knockdown, suggesting that NIK-mediated alternative activation of canonical NF-κB signaling is a key driver of pathological inflammatory activation of ECs. Targeting NIK could thus provide a novel approach for treating chronic inflammatory diseases.
Collapse
Affiliation(s)
- Paulina Kucharzewska
- Respiratory, Inflammation and Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| | - Chrissta X Maracle
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Kim C M Jeucken
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jan Piet van Hamburg
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Elisabeth Israelsson
- Respiratory, Inflammation and Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| | - Mark Furber
- Respiratory, Inflammation and Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Henric K Olsson
- Respiratory, Inflammation and Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, SE-431 83 Mölndal, Sweden
| |
Collapse
|
23
|
McKelvey KJ, Millier MJ, Doyle TC, Stamp LK, Highton J, Hessian PA. Co-expression of CD21L and IL17A defines a subset of rheumatoid synovia, characterised by large lymphoid aggregates and high inflammation. PLoS One 2018; 13:e0202135. [PMID: 30114200 PMCID: PMC6095528 DOI: 10.1371/journal.pone.0202135] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 07/27/2018] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE To determine whether the expression of IL17A and CD21L genes in inflamed rheumatoid synovia is associated with the neogenesis of ectopic lymphoid follicle-like structures (ELS), and if this aids the stratification of rheumatoid inflammation and thereby distinguishes patients with rheumatoid arthritis that might be responsive to specific targeted biologic therapies. METHODS Expression of IL17A and CD21L genes was assessed by RT-PCR, qRT-PCR and dPCR in synovia from 54 patients with rheumatoid arthritis. A subset of synovia (n = 30) was assessed by immunohistology for the presence of CD20+ B-lymphocytes and size of CD20+ B-lymphocyte aggregates as indicated by maximum radial cell count. The molecular profiles of six IL17A+/CD21L+ and six IL17A-/CD21L- synovia were determined by complementary DNA microarray analysis. RESULTS By RT-PCR, 26% of synovia expressed IL17A and 52% expressed CD21L. This provided the basis for distinguishing four subgroups of rheumatoid synovia: IL17A+/CD21L+ (18.5% of synovia), IL17A+/CD21L- (7.5%), IL17A-/CD21L+ (33.3%) and IL17A-/CD21L- (40.7%). While the subgroups did not predict clinical outcome measures, comparisons between the synovial subgroups revealed the IL17A+/CD21L+ subgroup had significantly larger CD20+ B-lymphocyte aggregates (P = 0.007) and a gene expression profile skewed toward B-cell- and antibody-mediated immunity. In contrast, genes associated with bone and cartilage remodelling were prominent in IL17A-/CD21L- synovia. CONCLUSIONS Rheumatoid synovia can be subdivided on the basis of IL17A and CD21L gene expression. Ensuing molecular subgroups do not predict clinical outcome for patients but highlight high inflammation and the predominance of B-lymphocyte mediated mechanisms operating in IL17A+/CD21L+ synovia. This may provide a rationale for more refined therapeutic selection due to the distinct molecular profiles associated with IL17A+/CD21L+ and IL17A-/CD21L- rheumatoid synovia.
Collapse
Affiliation(s)
- Kelly J. McKelvey
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Melanie J. Millier
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Terence C. Doyle
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Lisa K. Stamp
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - John Highton
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Paul A. Hessian
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| |
Collapse
|
24
|
Radke J, Koll R, Preuße C, Pehl D, Todorova K, Schönemann C, Allenbach Y, Aronica E, de Visser M, Heppner FL, Weis J, Doostkam S, Maisonobe T, Benveniste O, Goebel HH, Stenzel W. Architectural B-cell organization in skeletal muscle identifies subtypes of dermatomyositis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2018. [PMID: 29520367 PMCID: PMC5840889 DOI: 10.1212/nxi.0000000000000451] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Objective To study the B-cell content, organization, and existence of distinct B-cell subpopulations in relation to the expression of type 1 interferon signature related genes in dermatomyositis (DM). Methods Evaluation of skeletal muscle biopsies from patients with adult DM (aDM) and juvenile DM (jDM) by histology, immunohistochemistry, electron microscopy, and quantitative reverse-transcription PCR. Results We defined 3 aDM subgroups—classic (containing occasional B cells without clusters), B-cell–rich, and follicle-like aDM—further elucidating IM B-lymphocyte maturation and immunity. The quantity of B cells and formation of ectopic lymphoid structures in a subset of patients with aDM were associated with a specific profile of cytokines and chemokines involved in lymphoid neogenesis. Levels of type 1 interferon signature related gene expression paralleled B-cell content and architectural organization and link B-cell immunity to the interferon type I signature. Conclusion These data corroborate the important role of B cells in DM, highlighting the direct link between humoral mechanisms as key players in B-cell immunity and the role of type I interferon–related immunity.
Collapse
Affiliation(s)
- Josefine Radke
- Author affiliations are provided at the end of the article
| | - Randi Koll
- Author affiliations are provided at the end of the article
| | - Corinna Preuße
- Author affiliations are provided at the end of the article
| | - Debora Pehl
- Author affiliations are provided at the end of the article
| | | | | | - Yves Allenbach
- Author affiliations are provided at the end of the article
| | | | | | | | - Joachim Weis
- Author affiliations are provided at the end of the article
| | | | | | | | | | - Werner Stenzel
- Author affiliations are provided at the end of the article
| |
Collapse
|
25
|
Dendritic cell recruitment and activation in autoimmunity. J Autoimmun 2017; 85:126-140. [DOI: 10.1016/j.jaut.2017.07.012] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 07/26/2017] [Indexed: 12/11/2022]
|
26
|
Kim EY, Moudgil KD. Immunomodulation of autoimmune arthritis by pro-inflammatory cytokines. Cytokine 2017; 98:87-96. [PMID: 28438552 PMCID: PMC5581685 DOI: 10.1016/j.cyto.2017.04.012] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/09/2017] [Accepted: 04/10/2017] [Indexed: 12/18/2022]
Abstract
Pro-inflammatory cytokines promote autoimmune inflammation and tissue damage, while anti-inflammatory cytokines help resolve inflammation and facilitate tissue repair. Over the past few decades, this general feature of cytokine-mediated events has offered a broad framework to comprehend the pathogenesis of autoimmune and other immune-mediated diseases, and to successfully develop therapeutic approaches for diseases such as rheumatoid arthritis (RA). Anti-tumor necrosis factor-α (TNF-α) therapy is a testimony in support of this endeavor. However, many patients with RA fail to respond to this or other biologics, and some patients may suffer unexpected aggravation of arthritic inflammation or other autoimmune effects. These observations combined with rapid advancements in immunology in regard to newer cytokines and T cell subsets have enforced a re-evaluation of the perceived pathogenic attribute of the pro-inflammatory cytokines. Studies conducted by others and us in experimental models of arthritis involving direct administration of IFN-γ or TNF-α; in vivo neutralization of the cytokine; the use of animals deficient in the cytokine or its receptor; and the impact of the cytokine or anti-cytokine therapy on defined T cell subsets have revealed paradoxical anti-inflammatory and immunoregulatory attributes of these two cytokines. Similar studies in other models of autoimmunity as well as limited studies in arthritis patients have also unveiled the disease-protective effects of these pro-inflammatory cytokines. A major mechanism in this regard is the altered balance between the pathogenic T helper 17 (Th17) and protective T regulatory (Treg) cells in favor of the latter. However, it is essential to consider that this aspect of the pro-inflammatory cytokines is context-dependent such that the dose and timing of intervention, the experimental model of the disease under study, and the differences in individual responsiveness can influence the final outcomes. Nevertheless, the realization that pro-inflammatory cytokines can also be immunoregulatory offers a new perspective in fully understanding the pathogenesis of autoimmune diseases and in designing better therapies for controlling them.
Collapse
Affiliation(s)
- Eugene Y Kim
- Department of Pharmaceutical Sciences, School of Pharmacy, Washington State University, Spokane, WA, USA
| | - Kamal D Moudgil
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Division of Rheumatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
27
|
Al-Soudi A, Kaaij MH, Tas SW. Endothelial cells: From innocent bystanders to active participants in immune responses. Autoimmun Rev 2017; 16:951-962. [PMID: 28698091 DOI: 10.1016/j.autrev.2017.07.008] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 06/05/2017] [Indexed: 02/07/2023]
Abstract
The endothelium is crucially important for the delivery of oxygen and nutrients throughout the body under homeostatic conditions. However, it also contributes to pathology, including the initiation and perpetuation of inflammation. Understanding the function of endothelial cells (ECs) in inflammatory diseases and molecular mechanisms involved may lead to novel approaches to dampen inflammation and restore homeostasis. In this article, we discuss the various functions of ECs in inflammation with a focus on pathological angiogenesis, attraction of immune cells, antigen presentation, immunoregulatory properties and endothelial-to-mesenchymal transition (EndMT). We also review the current literature on approaches to target these processes in ECs to modulate immune responses and advance anti-inflammatory therapies.
Collapse
Affiliation(s)
- A Al-Soudi
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology & Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - M H Kaaij
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology & Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - S W Tas
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology & Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
28
|
Abstract
The nuclear factor-κB (NF-κB) family of transcription factors is activated by canonical and non-canonical signalling pathways, which differ in both signalling components and biological functions. Recent studies have revealed important roles for the non-canonical NF-κB pathway in regulating different aspects of immune functions. Defects in non-canonical NF-κB signalling are associated with severe immune deficiencies, whereas dysregulated activation of this pathway contributes to the pathogenesis of various autoimmune and inflammatory diseases. Here we review the signalling mechanisms and the biological function of the non-canonical NF-κB pathway. We also discuss recent progress in elucidating the molecular mechanisms regulating non-canonical NF-κB pathway activation, which may provide new opportunities for therapeutic strategies.
Collapse
Affiliation(s)
- Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, MD Anderson Cancer Center UT Heath Graduate School of Biomedical Sciences, 7455 Fannin Street, Box 902, Houston, Texas 77030, USA
| |
Collapse
|
29
|
|
30
|
Weinstein AM, Chen L, Brzana EA, Patil PR, Taylor JL, Fabian KL, Wallace CT, Jones SD, Watkins SC, Lu B, Stroncek DF, Denning TL, Fu YX, Cohen PA, Storkus WJ. Tbet and IL-36γ cooperate in therapeutic DC-mediated promotion of ectopic lymphoid organogenesis in the tumor microenvironment. Oncoimmunology 2017; 6:e1322238. [PMID: 28680760 DOI: 10.1080/2162402x.2017.1322238] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 12/31/2022] Open
Abstract
We have previously reported that direct injection of dendritic cells (DC) engineered to express the Type-1 transactivator Tbet (i.e., DC.Tbet) into murine tumors results in antitumor efficacy in association with the development of structures resembling tertiary lymphoid organs (TLO) in the tumor microenvironment (TME). These TLO contained robust infiltrates of B cells, DC, NK cells, and T cells in proximity to PNAd+ blood vessels; however, they were considered incomplete, since the recruited B cells failed to organize into classic germinal center-like structures. We now report that antitumor efficacy and TLO-inducing capacity of DC.Tbet-based i.t. therapy is operational in peripheral lymph node-deficient LTA-/- mice, and that it is highly dependent upon a direct Tbet target gene product, IL-36γ/IL-1F9. Intratumoral DC.Tbet fails to provide protection to tumor-bearing IL-36R-/- hosts, or to tumor-bearing wild-type recipient mice co-administered rmIL-1F5/IL-36RN, a natural IL-36R antagonist. Remarkably, the injection of tumors with DC engineered to secrete a bioactive form of mIL-36γ (DC.IL36γ) also initiated therapeutic TLO and slowed tumor progression in vivo. Furthermore, DC.IL36γ cells strongly upregulated their expression of Tbet, suggesting that Tbet and IL-36γ cooperate to reinforce each other's expression in DC, rendering them competent to promote TLO formation in an "immunologically normalized," therapeutic TME.
Collapse
Affiliation(s)
- Aliyah M Weinstein
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
| | - Lu Chen
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
| | | | | | | | - Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
| | - Callen T Wallace
- Department of Cell Biology and Physiology, UPSOM, Pittsburgh, PA, USA
| | | | - Simon C Watkins
- Department of Cell Biology and Physiology, UPSOM, Pittsburgh, PA, USA
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA
| | - David F Stroncek
- Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, PA, USA
| | - Timothy L Denning
- Center for Inflammation, Immunity & Infection at Georgia State University, Atlanta, GA, USA
| | - Yang-Xin Fu
- Departments of Pathology and Immunology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Peter A Cohen
- Department of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine (UPSOM), Pittsburgh, PA, USA.,Department of Dermatology, UPSOM, Pittsburgh, PA, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Bioengineering, UPSOM, Pittsburgh, PA, USA.,University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
31
|
Carmona-Rivera C, Carlucci PM, Moore E, Lingampalli N, Uchtenhagen H, James E, Liu Y, Bicker KL, Wahamaa H, Hoffmann V, Catrina AI, Thompson P, Buckner JH, Robinson WH, Fox DA, Kaplan MJ. Synovial fibroblast-neutrophil interactions promote pathogenic adaptive immunity in rheumatoid arthritis. Sci Immunol 2017. [PMID: 28649674 DOI: 10.1126/sciimmunol.aag3358] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Rheumatoid arthritis (RA) is characterized by synovial joint inflammation and by development of pathogenic humoral and cellular autoimmunity to citrullinated proteins. Neutrophil extracellular traps (NETs) are a source of citrullinated autoantigens and activate RA synovial fibroblasts (FLS), cells crucial in joint damage. We investigated the molecular mechanisms by which NETs promote proinflammatory phenotypes in FLS, and whether these interactions generate pathogenic anti-citrulline adaptive immune responses. NETs containing citrullinated peptides are internalized by FLS through a RAGE-TLR9 pathway promoting FLS inflammatory phenotype and their upregulation of MHC class II. Once internalized, arthritogenic NET-peptides are loaded into FLS MHC class II and presented to Ag-specific T cells. HLADRB1*0401 transgenic mice immunized with mouse FLS loaded with NETs develop antibodies specific to citrullinated forms of relevant RA autoantigens implicated in RA pathogenesis as well as cartilage damage. These results implicate FLS as mediators in RA pathogenesis, through the internalization and presentation of NET citrullinated peptides to the adaptive immune system leading to pathogenic autoimmunity and cartilage damage.
Collapse
Affiliation(s)
- Carmelo Carmona-Rivera
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Philip M Carlucci
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Erica Moore
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nithya Lingampalli
- VA Palo Alto Health Care System, Palo Alto, CA 94304, USA and the Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hannes Uchtenhagen
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Eddie James
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Yudong Liu
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kevin L Bicker
- Department of Chemistry, Middle Tennessee State University, 1301 E. Main St., Murfreesboro, TN 37132, USA
| | - Heidi Wahamaa
- Department of Medicine, Solna, Karolinska University Hospital, Stockholm S17176, Sweden
| | - Victoria Hoffmann
- Division of Veterinary Resources, Office of the Director, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anca Irinel Catrina
- Department of Medicine, Solna, Karolinska University Hospital, Stockholm S17176, Sweden
| | - PaulR Thompson
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jane H Buckner
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - William H Robinson
- VA Palo Alto Health Care System, Palo Alto, CA 94304, USA and the Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David A Fox
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
32
|
Flores-Borja F, Irshad S, Gordon P, Wong F, Sheriff I, Tutt A, Ng T. Crosstalk between Innate Lymphoid Cells and Other Immune Cells in the Tumor Microenvironment. J Immunol Res 2016; 2016:7803091. [PMID: 27882334 PMCID: PMC5110869 DOI: 10.1155/2016/7803091] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/20/2016] [Accepted: 08/30/2016] [Indexed: 12/26/2022] Open
Abstract
Our knowledge and understanding of the tumor microenvironment (TME) have been recently expanded with the recognition of the important role of innate lymphoid cells (ILC). Three different groups of ILC have been described based on their ability to produce cytokines that mediate the interactions between innate and adaptive immune cells in a variety of immune responses in infection, allergy, and autoimmunity. However, recent evidence from experimental models and clinical studies has demonstrated that ILC contribute to the mechanisms that generate suppressive or tolerant environments that allow tumor regression or progression. Defining the complex network of interactions and crosstalk of ILC with other immune cells and understanding the specific contributions of each type of ILC leading to tumor development will allow the manipulation of their function and will be important to develop new interventions and therapeutic strategies.
Collapse
Affiliation(s)
- Fabian Flores-Borja
- Breast Cancer Now Research Unit, Division of Cancer Studies, Guy's Hospital, King's College London School of Medicine, London SE1 9RT, UK
| | - Sheeba Irshad
- Breast Cancer Now Research Unit, Division of Cancer Studies, Guy's Hospital, King's College London School of Medicine, London SE1 9RT, UK
| | - Peter Gordon
- Breast Cancer Now Research Unit, Division of Cancer Studies, Guy's Hospital, King's College London School of Medicine, London SE1 9RT, UK
| | - Felix Wong
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, King's College London, Guy's Medical School Campus, London SE1 1ULK, UK
| | - Ibrahim Sheriff
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, King's College London, Guy's Medical School Campus, London SE1 1ULK, UK
| | - Andrew Tutt
- Breast Cancer Now Research Unit, Division of Cancer Studies, Guy's Hospital, King's College London School of Medicine, London SE1 9RT, UK
- Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London SW3 6JB, UK
| | - Tony Ng
- Breast Cancer Now Research Unit, Division of Cancer Studies, Guy's Hospital, King's College London School of Medicine, London SE1 9RT, UK
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, King's College London, Guy's Medical School Campus, London SE1 1ULK, UK
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6DD, UK
| |
Collapse
|
33
|
Coutant F, Miossec P. Altered dendritic cell functions in autoimmune diseases: distinct and overlapping profiles. Nat Rev Rheumatol 2016; 12:703-715. [DOI: 10.1038/nrrheum.2016.147] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
34
|
Serafini B, Rosicarelli B, Veroni C, Zhou L, Reali C, Aloisi F. RORγt Expression and Lymphoid Neogenesis in the Brain of Patients with Secondary Progressive Multiple Sclerosis. J Neuropathol Exp Neurol 2016; 75:877-88. [PMID: 27413074 DOI: 10.1093/jnen/nlw063] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Indexed: 12/16/2022] Open
Abstract
Ectopic B-cell follicle-like structures (ELS) are found in the meninges of patients with secondary progressive multiple sclerosis (SPMS). Because cells expressing the transcriptional regulator retinoic acid receptor-related orphan receptor-γt (RORγt) and producing interleukin 17 (IL17), e.g. T helper 17 cells and lymphoid tissue inducer (LTi) cells, have been implicated in the formation of ELS, we studied RORγt and IL17 expression in brain tissue from patients with SPMS an assessed their relationships to immune infiltrates and meningeal ELS. By immunohistochemistry, small numbers of RORγt-positive cells were detected in the meninges of 6 of 12 SPMS cases analyzed. RORγt-positive cells were localized in B-cell follicles or aggregates and nearby diffuse meningeal infiltrates, and predominantly co-expressed CD3. Only a few RORγt-positive, CD3-negative cells were observed, suggesting the presence of group 3 innate lymphoid cells, which comprise the LTi cell subset. Some IL17-positive cells, co-expressing in part RORγt and predominantly CD3, were found in meningeal B-cell follicles from 4 SPMS cases. Rare RORγt-positive and IL17-positive cells were detected in white matter. Gene expression analysis of laser dissected meningeal infiltrates and white matter lesions confirmed low frequencies and virtual absence of RORγt and IL17 signals, respectively. Thus, there is selective migration or survival of RORγt-positive cells in MS patient meninges and an association of these cells with ELS.
Collapse
Affiliation(s)
- Barbara Serafini
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Barbara Rosicarelli
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Caterina Veroni
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Ling Zhou
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Camilla Reali
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Francesca Aloisi
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR).
| |
Collapse
|
35
|
Padera TP, Meijer EFJ, Munn LL. The Lymphatic System in Disease Processes and Cancer Progression. Annu Rev Biomed Eng 2016; 18:125-58. [PMID: 26863922 DOI: 10.1146/annurev-bioeng-112315-031200] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Advances in our understanding of the structure and function of the lymphatic system have made it possible to identify its role in a variety of disease processes. Because it is involved not only in fluid homeostasis but also in immune cell trafficking, the lymphatic system can mediate and ultimately alter immune responses. Our rapidly increasing knowledge of the molecular control of the lymphatic system will inevitably lead to new and effective therapies for patients with lymphatic dysfunction. In this review, we discuss the molecular and physiological control of lymphatic vessel function and explore how the lymphatic system contributes to many disease processes, including cancer and lymphedema.
Collapse
Affiliation(s)
- Timothy P Padera
- Edwin L. Steele Laboratories, Department of Radiation Oncology, and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114;
| | - Eelco F J Meijer
- Edwin L. Steele Laboratories, Department of Radiation Oncology, and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114;
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114;
| |
Collapse
|