1
|
Wang Z, Cheng Y, Fan J, Luo R, Xu G, Ge S. Deletion of lymphotoxin-β receptor (LTβR) protects against acute kidney injury by PPARα pathway. Mol Med 2024; 30:254. [PMID: 39707217 DOI: 10.1186/s10020-024-01026-z] [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: 09/18/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Recent data has shown a considerable advancement in understanding the role of lymphotoxin-β receptor (LTβR) in inflammation. However, the functions and underlying mechanisms of LTβR in acute kidney injury (AKI) remain largely unknown. METHODS AKI was induced in mice by renal ischemia-reperfusion (I/R). HK-2 cells and primary renal tubular epithelial cells (RTECs) were subjected to hypoxia/reoxygenation (H/R) injury. The effects of LTβR depletion were examined in mice, as well as primary RTECs. Bone marrow chimeric mice was generated to determine whether the involvement of LTβR expression by parenchymal cells or bone marrow derived cells contributes to renal injury during AKI. RNA sequencing techniques were employed to investigate the mechanism via which LTβR signaling provides protection against I/R-induced AKI RESULTS: LTβR expression was downregulated both in vivo and in vitro models of AKI. Moreover, depletion of LTβR decreased renal damage and inflammation in I/R-induced AKI. We also found that LTβR deficient mice engrafted with wild type bone marrow had significantly less tubular damage, implying that LTβR in renal parenchymal cells may play dominant role in I/R-induced AKI. RNA sequencing indicated that the protective effect of LTβR deletion was associated with activation of PPARα signaling. Furthermore, upregulation of PPARα was observed upon depletion of LTβR. PPARα inhibitor, GW6471, aggravated the tubular damage and inflammation in LTβR-/- mice following I/R injury. Then we further demonstrated that LTβR depletion down-regulated non-canonical NF-κB and Bax/Bcl-2 apoptosis pathway through PPARα. CONCLUSIONS Our results suggested that the LTβR/PPARα axis may be a potential therapeutic target for the treatment of AKI.
Collapse
Affiliation(s)
- Zufeng Wang
- Department of Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Yichun Cheng
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan, 430030, China
| | - Jiahe Fan
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan, 430030, China
| | - Ran Luo
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan, 430030, China
| | - Gang Xu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan, 430030, China.
| | - Shuwang Ge
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan, 430030, China.
| |
Collapse
|
2
|
Craig R, McIntosh K, Ho Ho K, McCulloch A, Riley C, Lawson C, Mackay SP, Paul A, Coats P, Plevin R. IL-1β stimulates a novel axis within the NFκB pathway in endothelial cells regulated by IKKα and TAK-1. Biochem Pharmacol 2024:116736. [PMID: 39710275 DOI: 10.1016/j.bcp.2024.116736] [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: 08/27/2024] [Revised: 12/11/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
In this study we examined the activation of the non-canonical NFκB signalling pathway in endothelial cells. In HUVECs, LIGHT stimulated a delayed induction of serine 866/870 p100 phosphorylation linked to p52 NFκB formation. Surprisingly, the canonical ligand, IL-1β, stimulated a rapid phosphorylation or p100 which was not associated with p52 formation. Inhibition of IKKα activity, using DN-IKKα adenovirus, IKKα siRNA or a novel first-in-class selective IKKα inhibitor, SU1261, revealed IL-1β induced p100 phosphorylation to be dependent on IKKα. In contrast, IKKβ inhibition was found to be without effect. The NIK inhibitor, CW15337, did not affect IL-1β induced p100 phosphorylation however, both p100 and pIKKα/β phosphorylation was substantially reduced by inhibition of the upstream kinase TAK-1, suggesting phosphorylation of p100 is mediated by IKKα from within the canonical NEMO/IKKβ /IKKα complex. IL-1β also stimulated a rapid increase in nuclear translocation of p52, which was not affected by NIK inhibition, suggesting a source of p52 independent of p100 processing. Inhibition of TAK-1 abolished p52 and p65 nuclear translocation in response to IL-1β. SiRNA deletion or inhibition with dominant-negative virus of IKKα activity partially reduced p52 translocation, however pharmacological inhibition of IKKα was without effect. Inhibition of IKKβ abolished both p52 and p65 translocation. Taken together these results show that IL-1β stimulates a novel IKKα -dependent axis within the non-canonical NFκB pathway in endothelial cells which is NIK-independent and regulated by TAK-1. However, this pathway is not primarily responsible for the early nuclear translocation of p52, which is dependent on IKKβ. Elucidation of both these new pathways may be significant for NFκB biology within the endothelium.
Collapse
Affiliation(s)
- Rachel Craig
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, Scotland, UK
| | - Kathryn McIntosh
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, Scotland, UK.
| | - Ka Ho Ho
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, Scotland, UK
| | - Ashley McCulloch
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, Scotland, UK
| | - Christopher Riley
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, Scotland, UK
| | - Christopher Lawson
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, Scotland, UK
| | - Simon P Mackay
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, Scotland, UK
| | - Andrew Paul
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, Scotland, UK
| | - Paul Coats
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, Scotland, UK
| | - Robin Plevin
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, Scotland, UK.
| |
Collapse
|
3
|
Cao Y, Yi Y, Han C, Shi B. NF-κB signaling pathway in tumor microenvironment. Front Immunol 2024; 15:1476030. [PMID: 39493763 PMCID: PMC11530992 DOI: 10.3389/fimmu.2024.1476030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
The genesis and progression of tumors are multifaceted processes influenced by genetic mutations within the tumor cells and the dynamic interplay with their surrounding milieu, which incessantly impacts the course of cancer. The tumor microenvironment (TME) is a complex and dynamic entity that encompasses not only the tumor cells but also an array of non-cancerous cells, signaling molecules, and the extracellular matrix. This intricate network is crucial in tumor progression, metastasis, and response to treatments. The TME is populated by diverse cell types, including immune cells, fibroblasts, endothelial cells, alongside cytokines and growth factors, all of which play roles in either suppressing or fostering tumor growth. Grasping the nuances of the interactions within the TME is vital for the advancement of targeted cancer therapies. Consequently, a thorough understanding of the alterations of TME and the identification of upstream regulatory targets have emerged as a research priority. NF-κB transcription factors, central to inflammation and innate immunity, are increasingly recognized for their significant role in cancer onset and progression. This review emphasizes the crucial influence of the NF-κB signaling pathway within the TME, underscoring its roles in the development and advancement of cancer. By examining the interactions between NF-κB and various components of the TME, targeting the NF-κB pathway appears as a promising cancer treatment approach.
Collapse
Affiliation(s)
- Yaning Cao
- Department of Blood Transfusion, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu, China
| | - Yanan Yi
- Department of Laboratory Medicine, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Chongxu Han
- Department of Laboratory Medicine, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Bingwei Shi
- Department of Blood Transfusion, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu, China
| |
Collapse
|
4
|
Wang X, Zhang T, Zheng B, Lu Y, Liang Y, Xu G, Zhao L, Tao Y, Song Q, You H, Hu H, Li X, Sun K, Li T, Zhang Z, Wang J, Lan X, Pan D, Fu YX, Yue B, Zheng H. Lymphotoxin-β promotes breast cancer bone metastasis colonization and osteolytic outgrowth. Nat Cell Biol 2024; 26:1597-1612. [PMID: 39147874 DOI: 10.1038/s41556-024-01478-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 07/11/2024] [Indexed: 08/17/2024]
Abstract
Bone metastasis is a lethal consequence of breast cancer. Here we used single-cell transcriptomics to investigate the molecular mechanisms underlying bone metastasis colonization-the rate-limiting step in the metastatic cascade. We identified that lymphotoxin-β (LTβ) is highly expressed in tumour cells within the bone microenvironment and this expression is associated with poor bone metastasis-free survival. LTβ promotes tumour cell colonization and outgrowth in multiple breast cancer models. Mechanistically, tumour-derived LTβ activates osteoblasts through nuclear factor-κB2 signalling to secrete CCL2/5, which facilitates tumour cell adhesion to osteoblasts and accelerates osteoclastogenesis, leading to bone metastasis progression. Blocking LTβ signalling with a decoy receptor significantly suppressed bone metastasis in vivo, whereas clinical sample analysis revealed significantly higher LTβ expression in bone metastases than in primary tumours. Our findings highlight LTβ as a bone niche-induced factor that promotes tumour cell colonization and osteolytic outgrowth and underscore its potential as a therapeutic target for patients with bone metastatic disease.
Collapse
Affiliation(s)
- Xuxiang Wang
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Tengjiang Zhang
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Bingxin Zheng
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Youxue Lu
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Yong Liang
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Guoyuan Xu
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Luyang Zhao
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Yuwei Tao
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Qianhui Song
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Huiwen You
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Haitian Hu
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Xuan Li
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Keyong Sun
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Tianqi Li
- School of Life Sciences and Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, China
| | - Zian Zhang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianbin Wang
- School of Life Sciences and Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, China
| | - Xun Lan
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Deng Pan
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Yang-Xin Fu
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Bin Yue
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Hanqiu Zheng
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China.
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
5
|
Riley C, Ammar U, Alsfouk A, Anthony NG, Baiget J, Berretta G, Breen D, Huggan J, Lawson C, McIntosh K, Plevin R, Suckling CJ, Young LC, Paul A, Mackay SP. Design and Synthesis of Novel Aminoindazole-pyrrolo[2,3- b]pyridine Inhibitors of IKKα That Selectively Perturb Cellular Non-Canonical NF-κB Signalling. Molecules 2024; 29:3515. [PMID: 39124921 PMCID: PMC11314561 DOI: 10.3390/molecules29153515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/11/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
The inhibitory-kappaB kinases (IKKs) IKKα and IKKβ play central roles in regulating the non-canonical and canonical NF-κB signalling pathways. Whilst the proteins that transduce the signals of each pathway have been extensively characterised, the clear dissection of the functional roles of IKKα-mediated non-canonical NF-κB signalling versus IKKβ-driven canonical signalling remains to be fully elucidated. Progress has relied upon complementary molecular and pharmacological tools; however, the lack of highly potent and selective IKKα inhibitors has limited advances. Herein, we report the development of an aminoindazole-pyrrolo[2,3-b]pyridine scaffold into a novel series of IKKα inhibitors. We demonstrate high potency and selectivity against IKKα over IKKβ in vitro and explain the structure-activity relationships using structure-based molecular modelling. We show selective target engagement with IKKα in the non-canonical NF-κB pathway for both U2OS osteosarcoma and PC-3M prostate cancer cells by employing isoform-related pharmacodynamic markers from both pathways. Two compounds (SU1261 [IKKα Ki = 10 nM; IKKβ Ki = 680 nM] and SU1349 [IKKα Ki = 16 nM; IKKβ Ki = 3352 nM]) represent the first selective and potent pharmacological tools that can be used to interrogate the different signalling functions of IKKα and IKKβ in cells. Our understanding of the regulatory role of IKKα in various inflammatory-based conditions will be advanced using these pharmacological agents.
Collapse
Affiliation(s)
- Christopher Riley
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Usama Ammar
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Aisha Alsfouk
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Nahoum G. Anthony
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Jessica Baiget
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Giacomo Berretta
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - David Breen
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Judith Huggan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Christopher Lawson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Kathryn McIntosh
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Robin Plevin
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Colin J. Suckling
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Louise C. Young
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Andrew Paul
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Simon P. Mackay
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| |
Collapse
|
6
|
Cully MD, Nolte JE, Patel A, Vaughan AE, May MJ. Loss of Lymphatic IKKα Disrupts Lung Immune Homeostasis, Drives BALT Formation, and Protects against Influenza. Immunohorizons 2024; 8:478-491. [PMID: 39007717 PMCID: PMC11294277 DOI: 10.4049/immunohorizons.2400047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024] Open
Abstract
IκB kinase (IKK)α controls noncanonical NF-κB signaling required for lymphoid organ development. We showed previously that lymph node formation is ablated in IkkαLyve-1 mice constitutively lacking IKKα in lymphatic endothelial cells (LECs). We now reveal that loss of IKKα in LECs leads to the formation of BALT in the lung. Tertiary lymphoid structures appear only in the lungs of IkkαLyve-1 mice and are not present in any other tissues, and these highly organized BALT structures form after birth and in the absence of inflammation. Additionally, we show that IkkαLyve-1 mice challenged with influenza A virus (IAV) exhibit markedly improved survival and reduced weight loss compared with littermate controls. Importantly, we determine that the improved morbidity and mortality of IkkαLyve-1 mice is independent of viral load and rate of clearance because both mice control and clear IAV infection similarly. Instead, we show that IFN-γ levels are decreased, and infiltration of CD8 T cells and monocytes into IkkαLyve-1 lungs is reduced. We conclude that ablating IKKα in LECs promotes BALT formation and reduces the susceptibility of IkkαLyve-1 mice to IAV infection through a decrease in proinflammatory stimuli.
Collapse
Affiliation(s)
- Michelle D. Cully
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Julianne E. Nolte
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Athena Patel
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Andrew E. Vaughan
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Michael J. May
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| |
Collapse
|
7
|
Walitt B, Singh K, LaMunion SR, Hallett M, Jacobson S, Chen K, Enose-Akahata Y, Apps R, Barb JJ, Bedard P, Brychta RJ, Buckley AW, Burbelo PD, Calco B, Cathay B, Chen L, Chigurupati S, Chen J, Cheung F, Chin LMK, Coleman BW, Courville AB, Deming MS, Drinkard B, Feng LR, Ferrucci L, Gabel SA, Gavin A, Goldstein DS, Hassanzadeh S, Horan SC, Horovitz SG, Johnson KR, Govan AJ, Knutson KM, Kreskow JD, Levin M, Lyons JJ, Madian N, Malik N, Mammen AL, McCulloch JA, McGurrin PM, Milner JD, Moaddel R, Mueller GA, Mukherjee A, Muñoz-Braceras S, Norato G, Pak K, Pinal-Fernandez I, Popa T, Reoma LB, Sack MN, Safavi F, Saligan LN, Sellers BA, Sinclair S, Smith B, Snow J, Solin S, Stussman BJ, Trinchieri G, Turner SA, Vetter CS, Vial F, Vizioli C, Williams A, Yang SB, Nath A. Deep phenotyping of post-infectious myalgic encephalomyelitis/chronic fatigue syndrome. Nat Commun 2024; 15:907. [PMID: 38383456 PMCID: PMC10881493 DOI: 10.1038/s41467-024-45107-3] [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: 06/24/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024] Open
Abstract
Post-infectious myalgic encephalomyelitis/chronic fatigue syndrome (PI-ME/CFS) is a disabling disorder, yet the clinical phenotype is poorly defined, the pathophysiology is unknown, and no disease-modifying treatments are available. We used rigorous criteria to recruit PI-ME/CFS participants with matched controls to conduct deep phenotyping. Among the many physical and cognitive complaints, one defining feature of PI-ME/CFS was an alteration of effort preference, rather than physical or central fatigue, due to dysfunction of integrative brain regions potentially associated with central catechol pathway dysregulation, with consequences on autonomic functioning and physical conditioning. Immune profiling suggested chronic antigenic stimulation with increase in naïve and decrease in switched memory B-cells. Alterations in gene expression profiles of peripheral blood mononuclear cells and metabolic pathways were consistent with cellular phenotypic studies and demonstrated differences according to sex. Together these clinical abnormalities and biomarker differences provide unique insight into the underlying pathophysiology of PI-ME/CFS, which may guide future intervention.
Collapse
Affiliation(s)
- Brian Walitt
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Komudi Singh
- National Heart, Lung and Blood Institute (NHLBI), Bethesda, MD, USA
| | - Samuel R LaMunion
- National Institute of Diabetes, Digestion, and Kidney Disease (NIDDK), Bethesda, MD, USA
| | - Mark Hallett
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Steve Jacobson
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Kong Chen
- National Institute of Diabetes, Digestion, and Kidney Disease (NIDDK), Bethesda, MD, USA
| | | | - Richard Apps
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, MD, USA
| | | | - Patrick Bedard
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Robert J Brychta
- National Institute of Diabetes, Digestion, and Kidney Disease (NIDDK), Bethesda, MD, USA
| | | | - Peter D Burbelo
- National Institute of Dental and Craniofacial Research (NIDCR), Bethesda, MD, USA
| | - Brice Calco
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Brianna Cathay
- Texas A&M School of Engineering Medicine, College Station, TX, USA
| | - Li Chen
- Affiliated Hospital of North Sichuan Medical College, Sichuan, China
| | - Snigdha Chigurupati
- George Washington University Hospital, District of Columbia, Washington, DC, USA
| | - Jinguo Chen
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, MD, USA
| | - Foo Cheung
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, MD, USA
| | | | | | - Amber B Courville
- National Institute of Diabetes, Digestion, and Kidney Disease (NIDDK), Bethesda, MD, USA
| | | | | | | | | | - Scott A Gabel
- National Institute of Environmental Health Sciences (NIEHS), Chapel Hill, NC, USA
| | - Angelique Gavin
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - David S Goldstein
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | | | - Sean C Horan
- Sidney Kimmel Medical College, Philadelphia, PA, USA
| | - Silvina G Horovitz
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Kory R Johnson
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Anita Jones Govan
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Kristine M Knutson
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Joy D Kreskow
- National Institute of Nursing Research (NINR), Bethesda, MD, USA
| | - Mark Levin
- National Heart, Lung and Blood Institute (NHLBI), Bethesda, MD, USA
| | - Jonathan J Lyons
- National Institute of Allergy and Infectious Disease (NIAID), Bethesda, MD, USA
| | - Nicholas Madian
- National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD, USA
| | - Nasir Malik
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Andrew L Mammen
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), Bethesda, MD, USA
| | | | - Patrick M McGurrin
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | | | - Ruin Moaddel
- National Institute of Aging (NIA), Baltimore, MD, USA
| | - Geoffrey A Mueller
- National Institute of Environmental Health Sciences (NIEHS), Chapel Hill, NC, USA
| | - Amrita Mukherjee
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, MD, USA
| | - Sandra Muñoz-Braceras
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), Bethesda, MD, USA
| | - Gina Norato
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Katherine Pak
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), Bethesda, MD, USA
| | - Iago Pinal-Fernandez
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), Bethesda, MD, USA
| | - Traian Popa
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Lauren B Reoma
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Michael N Sack
- National Heart, Lung and Blood Institute (NHLBI), Bethesda, MD, USA
| | - Farinaz Safavi
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
- National Institute of Allergy and Infectious Disease (NIAID), Bethesda, MD, USA
| | - Leorey N Saligan
- National Institute of Nursing Research (NINR), Bethesda, MD, USA
| | - Brian A Sellers
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, MD, USA
| | | | - Bryan Smith
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Joseph Snow
- National Institute of Mental Health (NIMH), Bethesda, MD, USA
| | | | - Barbara J Stussman
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
- National Center for Complementary and Integrative Health (NCCIH), Bethesda, MD, USA
| | | | | | | | - Felipe Vial
- Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Carlotta Vizioli
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA
| | - Ashley Williams
- Oakland University William Beaumont School of Medicine, Rochester, NY, USA
| | | | - Avindra Nath
- National Institute of Neurological Diseases and Stroke (NINDS), Bethesda, MD, USA.
| |
Collapse
|
8
|
Jiang Y, Zhang J, Shi C, Li X, Jiang Y, Mao R. NF- κB: a mediator that promotes or inhibits angiogenesis in human diseases? Expert Rev Mol Med 2023; 25:e25. [PMID: 37503730 DOI: 10.1017/erm.2023.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The nuclear factor of κ-light chain of enhancer-activated B cells (NF-κB) signaling pathway, which is conserved in invertebrates, plays a significant role in human diseases such as inflammation-related diseases and carcinogenesis. Angiogenesis refers to the growth of new capillary vessels derived from already existing capillaries and postcapillary venules. Maintaining normal angiogenesis and effective vascular function is a prerequisite for the stability of organ tissue function, and abnormal angiogenesis often leads to a variety of diseases. It has been suggested that NK-κB signalling molecules under pathological conditions play an important role in vascular differentiation, proliferation, apoptosis and tumourigenesis by regulating the transcription of multiple target genes. Many NF-κB inhibitors are being tested in clinical trials for cancer treatment and their effect on angiogenesis is summarised. In this review, we will summarise the role of NF-κB signalling in various neovascular diseases, especially in tumours, and explore whether NF-κB can be used as an attack target or activation medium to inhibit tumour angiogenesis.
Collapse
Affiliation(s)
- Yijing Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Jie Zhang
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, 30Tongyang North Road, Pingchao Town, Nantong 226361, Jiangsu, People's Republic of China
| | - Conglin Shi
- Department of Pathogenic Biology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Xingjuan Li
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Yongying Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| |
Collapse
|
9
|
McIntosh K, Khalaf YH, Craig R, West C, McCulloch A, Waghmare A, Lawson C, Chan EYW, Mackay S, Paul A, Plevin R. IL-1β stimulates a novel, IKKα -dependent, NIK -independent activation of non-canonical NFκB signalling. Cell Signal 2023; 107:110684. [PMID: 37080443 DOI: 10.1016/j.cellsig.2023.110684] [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: 10/04/2022] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
In this study, we examined the activation of non-canonical nuclear factor Kappa B (NFκB) signalling in U2OS cells, a cellular metastatic bone cancer model. Whilst Lymphotoxin α1β2 (LTα1β2) stimulated the expected slow, delayed, sustained activation of serine 866/870 p100 phosphorylation and increased cellular expression of p52 NFκB, we found that canonical agonists, Interleukin-1β (IL-1β) and also Tumour necrosis factor-α (TNFα) generated a rapid transient increase in pp100, which was maximal by 15-30 min. This rapid phosphorylation was also observed in other cells types, such as DU145 and HCAECs suggesting the phenomenon is universal. IKKα deletion using CRISPR/Cas9 revealed an IKKα-dependent mechanism for serine 866/870 and additionally serine 872 p100 phosphorylation for both IL-1β and LTα1β2. In contrast, knockdown of IKKβ using siRNA or pharmacological inhibition of IKKβ activity was without effect on p100 phosphorylation. Pre-incubation of cells with the NFκB inducing-kinase (NIK) inhibitor, CW15337, had no effect on IL-1β induced phosphorylation of p100 however, the response to LTα1β2 was virtually abolished. Surprisingly IL-1β also stimulated p52 nuclear translocation as early as 60 min, this response and the concomitant p65 translocation was partially reduced by IKKα deletion. Furthermore, p52 nuclear translocation was unaffected by CW15337. In contrast, the response to LTα1β2 was essentially abolished by both IKKα deletion and CW15337. Taken together, these finding reveal novel forms of NFκB non-canonical signalling stimulated by ligands that activate the canonical NFκB pathway strongly such as IL-1β.
Collapse
Affiliation(s)
- Kathryn McIntosh
- Strathclyde Institute for Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE Scotland, UK.
| | - Yousif H Khalaf
- Strathclyde Institute for Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE Scotland, UK
| | - Rachel Craig
- Strathclyde Institute for Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE Scotland, UK
| | - Christopher West
- Strathclyde Institute for Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE Scotland, UK
| | - Ashley McCulloch
- Strathclyde Institute for Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE Scotland, UK
| | - Ajay Waghmare
- Strathclyde Institute for Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE Scotland, UK
| | - Christopher Lawson
- Strathclyde Institute for Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE Scotland, UK
| | - Edmond Y W Chan
- Department of Biomedical and Molecular sciences, Queens University, Botterell Hall, Room 563, 18 Stuart Street, Kingston, ON K7L 3N6, Canada
| | - Simon Mackay
- Strathclyde Institute for Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE Scotland, UK
| | - Andrew Paul
- Strathclyde Institute for Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE Scotland, UK
| | - Robin Plevin
- Strathclyde Institute for Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE Scotland, UK.
| |
Collapse
|
10
|
Aaron T, Laudermilch E, Benet Z, Ovando LJ, Chandran K, Fooksman D. TNF-α Limits Serological Memory by Disrupting the Bone Marrow Niche. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:595-608. [PMID: 36645344 PMCID: PMC9998356 DOI: 10.4049/jimmunol.2200053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 12/17/2022] [Indexed: 01/17/2023]
Abstract
Both infection and autoimmune disease can disrupt pre-existing Ab titers leading to diminished serological memory, yet the underlying mechanisms are not well understood. In this article, we report that TNF-α, an inflammatory cytokine, is a master regulator of the plasma cell (PC) niche in the bone marrow (BM). Acute rTNF-α treatment depletes previously existing Ab titers after vaccination by limiting PC occupancy or retention in the BM. Consistent with this phenomenon, mice lacking TNF-α signaling have elevated PC capacity in the BM and higher Ab titers. Using BM chimeric mice, we found that PC egress from the BM is regulated in a cell-extrinsic manner, by radiation-resistant cells via TNF-α receptor 1 signaling, leading to increased vascular permeability and CD138 downregulation on PCs. PC motility and egress in the BM are triggered within 6 h of recombinant TNF-α treatment. In addition to promoting egress, TNF-α signaling also prevented re-engraftment into the BM, leading to reduced PC survival. Although other inflammatory stimuli can promote PC egress, TNF-α signaling is necessary for limiting the PC capacity in the BM. Collectively, these data characterize how TNF-α-mediated inflammation attenuates the durability of serological memory and shapes the overall size and composition of the Ab-secreting cell pool in the BM.
Collapse
Affiliation(s)
- Tonya Aaron
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Ethan Laudermilch
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Zachary Benet
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Luis Jose Ovando
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - David Fooksman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
11
|
Jeucken KCM, van Rooijen CCN, Kan YY, Kocken LA, Jongejan A, van Steen ACI, van Buul JD, Olsson HK, van Hamburg JP, Tas SW. Differential Contribution of NF-κB Signaling Pathways to CD4+ Memory T Cell Induced Activation of Endothelial Cells. Front Immunol 2022; 13:860327. [PMID: 35769477 PMCID: PMC9235360 DOI: 10.3389/fimmu.2022.860327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 05/12/2022] [Indexed: 11/21/2022] Open
Abstract
Endothelial cells (ECs) are important contributors to inflammation in immune-mediated inflammatory diseases (IMIDs). In this study, we examined whether CD4+ memory T (Tm) cells can drive EC inflammatory responses. Human Tm cells produced ligands that induced inflammatory responses in human umbilical vein EC as exemplified by increased expression of inflammatory mediators including chemokines and adhesion molecules. NF-κB, a key regulator of EC activation, was induced by Tm cell ligands. We dissected the relative contribution of canonical and non-canonical NF-κB signaling to Tm induced EC responses using pharmacological small molecule inhibitors of IKKβ (iIKKβ) or NF-κB inducing kinase (iNIK). RNA sequencing revealed substantial overlap in IKKβ and NIK regulated genes (n=549) that were involved in inflammatory and immune responses, including cytokines (IL-1β, IL-6, GM-CSF) and chemokines (CXCL5, CXCL1). NIK regulated genes were more restricted, as 332 genes were uniquely affected by iNIK versus 749 genes by iIKKβ, the latter including genes involved in metabolism, proliferation and leukocyte adhesion (VCAM-1, ICAM-1). The functional importance of NIK and IKKβ in EC activation was confirmed by transendothelial migration assays with neutrophils, demonstrating stronger inhibitory effects of iIKKβ compared to iNIK. Importantly, iIKKβ – and to some extent iNIK - potentiated the effects of currently employed therapies for IMIDs, like JAK inhibitors and anti-IL-17 antibodies, on EC inflammatory responses. These data demonstrate that inhibition of NF-κB signaling results in modulation of Tm cell-induced EC responses and highlight the potential of small molecule NF-κB inhibitors as a novel treatment strategy to target EC inflammatory responses in IMIDs.
Collapse
Affiliation(s)
- Kim C. M. Jeucken
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Charlotte C. N. van Rooijen
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Yik Y. Kan
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Lotte A. Kocken
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Aldo Jongejan
- Department of Epidemiology and Data Science, Bioinformatics Laboratory, Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Abraham C. I. van Steen
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, Netherlands
| | - Jaap D. van Buul
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, Netherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), Section Molecular Cytology, Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, Amsterdam, Netherlands
| | - Henric K. Olsson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jan Piet van Hamburg
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Sander W. Tas
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Sander W. Tas,
| |
Collapse
|
12
|
Ko S, Lim J, Hong S. Functional characterization of a novel tumor necrosis factor gene (TNF-New) in rock bream (Oplegnathus fasciatus). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 127:104269. [PMID: 34600021 DOI: 10.1016/j.dci.2021.104269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/03/2021] [Accepted: 09/24/2021] [Indexed: 06/13/2023]
Abstract
The novel tumor necrosis factor (TNF-New or TNFN) gene has been identified only in teleost such as zebrafish, medaka (Oryzias latipes), fugu (Takifugu rubripes), and rainbow trout (Oncorhynchus mykiss). In this study, a putative TNFN gene in rock bream (named RB-TNFN) was cloned and its functional expression in the immune system was analyzed. Although it was previously reported to share a high degree of homology with mammalian lymphotoxin (LT)-β, in silico analysis revealed that RB-TNFN differed slightly from mammalian LT-β in its genomic structure, phylogenetic relationship, and predicted protein tertiary structure, whereas the genomic location of TNFN (immediately behind TNF-α) was the same as that of LT-β. In healthy rock bream, RB-TNFN gene expression was the highest in the liver and the lowest in the head kidney. In vitro, it was significantly upregulated in head kidney cells following polyinosinic:polycytidylic acid, concanavalin A, phytohemagglutinin, or calcium ionophore (CI) stimulation and in spleen cells by lipopolysaccharide (LPS), CI, and rock bream iridovirus (RBIV). In vivo, it was upregulated in the spleen, liver, and gut on day 1 and in the blood on day 3 following LPS injection, and in the blood, head kidney, and liver following RBIV vaccination. Post-RBIV infection, the vaccinated group showed a significantly higher TNFN gene expression in the head kidney and blood than the unvaccinated group. Treatment with recombinant TNFN protein (RB-rTNFN) resulted in significantly upregulated interleukin-1β expression in the head kidney, spleen, blood, liver, and peritoneal cells. It also enhanced IL-8 gene expression in the head kidney, blood, and peritoneal cells, and interferon γ gene expression in the gut and gills on day 1. TNFN and cyclo-oxygenase-2 gene expression was upregulated in peritoneal cells on day 3. Flow cytometry analysis revealed a significant increase in the peritoneal lymphocyte population after the intraperitoneal (i.p.) injection of RB-rTNFN. These results suggest that RB-TNFN mediated innate and adaptive immunity in rock bream.
Collapse
Affiliation(s)
- Sungjae Ko
- Department of Marine Biotechnology, Gangneung-Wonju National University, Gangneung, South Korea
| | - Jongwon Lim
- Department of Marine Biotechnology, Gangneung-Wonju National University, Gangneung, South Korea
| | - Suhee Hong
- East Coast Research institute of Life Science, Gangneung-Wonju National University, South Korea.
| |
Collapse
|
13
|
A20 undermines alternative NF-κB activity and expression of anti-apoptotic genes in Helicobacter pylori infection. Cell Mol Life Sci 2022; 79:102. [PMID: 35089437 PMCID: PMC8799570 DOI: 10.1007/s00018-022-04139-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/16/2021] [Accepted: 01/08/2022] [Indexed: 12/16/2022]
Abstract
A hallmark of infection by the pathogen Helicobacter pylori, which colonizes the human gastric epithelium, is the simultaneous activation of the classical and alternative nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathways, underlying inflammation and cell survival. Here, we report that the classical NF-κB target gene product A20 contributes to the negative regulation of alternative NF-κB signaling in gastric epithelial cells infected by H. pylori. Mechanistically, the de novo synthesized A20 protein interacts with tumor necrosis factor receptor-associated factor-interacting protein with forkhead-associated domain (TIFA) and thereby interferes with the association of TIFA with the NIK regulatory complex. We also show that alternative NF-κB activity contributes to the up-regulation of anti-apoptotic genes, such as baculoviral IAP repeat containing 2 (BIRC2), BIRC3 and B-cell lymphoma 2-related protein A1 (BCL2A1) in gastric epithelial cells. Furthermore, the observed over-expression of RelB in human gastric biopsies with type B gastritis and RelB-dependent suppression of apoptotic cell death emphasize an important role of the alternative NF-κB pathway in H. pylori infection.
Collapse
|
14
|
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
|
15
|
Piao W, Kasinath V, Saxena V, Lakhan R, Iyyathurai J, Bromberg JS. LTβR Signaling Controls Lymphatic Migration of Immune Cells. Cells 2021; 10:cells10040747. [PMID: 33805271 PMCID: PMC8065509 DOI: 10.3390/cells10040747] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
The pleiotropic functions of lymphotoxin (LT)β receptor (LTβR) signaling are linked to the control of secondary lymphoid organ development and structural maintenance, inflammatory or autoimmune disorders, and carcinogenesis. Recently, LTβR signaling in endothelial cells has been revealed to regulate immune cell migration. Signaling through LTβR is comprised of both the canonical and non-canonical-nuclear factor κB (NF-κB) pathways, which induce chemokines, cytokines, and cell adhesion molecules. Here, we focus on the novel functions of LTβR signaling in lymphatic endothelial cells for migration of regulatory T cells (Tregs), and specific targeting of LTβR signaling for potential therapeutics in transplantation and cancer patient survival.
Collapse
Affiliation(s)
- Wenji Piao
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (W.P.); (R.L.)
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (V.S.); (J.I.)
| | - Vivek Kasinath
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Vikas Saxena
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (V.S.); (J.I.)
| | - Ram Lakhan
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (W.P.); (R.L.)
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (V.S.); (J.I.)
| | - Jegan Iyyathurai
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (V.S.); (J.I.)
| | - Jonathan S. Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (W.P.); (R.L.)
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (V.S.); (J.I.)
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Correspondence: ; Tel.: +410-328-6430
| |
Collapse
|
16
|
Höpner SS, Raykova A, Radpour R, Amrein MA, Koller D, Baerlocher GM, Riether C, Ochsenbein AF. LIGHT/LTβR signaling regulates self-renewal and differentiation of hematopoietic and leukemia stem cells. Nat Commun 2021; 12:1065. [PMID: 33594067 PMCID: PMC7887212 DOI: 10.1038/s41467-021-21317-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/17/2021] [Indexed: 12/30/2022] Open
Abstract
The production of blood cells during steady-state and increased demand depends on the regulation of hematopoietic stem cell (HSC) self-renewal and differentiation. Similarly, the balance between self-renewal and differentiation of leukemia stem cells (LSCs) is crucial in the pathogenesis of leukemia. Here, we document that the TNF receptor superfamily member lymphotoxin-β receptor (LTβR) and its ligand LIGHT regulate quiescence and self-renewal of murine and human HSCs and LSCs. Cell-autonomous LIGHT/LTβR signaling on HSCs reduces cell cycling, promotes symmetric cell division and prevents primitive HSCs from exhaustion in serial re-transplantation experiments and genotoxic stress. LTβR deficiency reduces the numbers of LSCs and prolongs survival in a murine chronic myeloid leukemia (CML) model. Similarly, LIGHT/LTβR signaling in human G-CSF mobilized HSCs and human LSCs results in increased colony forming capacity in vitro. Thus, our results define LIGHT/LTβR signaling as an important pathway in the regulation of the self-renewal of HSCs and LSCs.
Collapse
MESH Headings
- Animals
- Antigens, CD34/metabolism
- Cell Cycle/drug effects
- Cell Cycle/genetics
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cell Self Renewal/drug effects
- Cell Self Renewal/genetics
- DNA Damage
- Fluorouracil/pharmacology
- Gene Expression Regulation, Leukemic/drug effects
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Lymphotoxin beta Receptor/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction/drug effects
- Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
- Mice
Collapse
Affiliation(s)
- S S Höpner
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Ana Raykova
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - R Radpour
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - M A Amrein
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - D Koller
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - G M Baerlocher
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - C Riether
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - A F Ochsenbein
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
17
|
Vanoni G, Ercolano G, Candiani S, Rutigliani M, Lanata M, Derré L, Marcenaro E, Schneider P, Romero P, Jandus C, Trabanelli S. Human primed ILCPs support endothelial activation through NF-κB signaling. eLife 2021; 10:e58838. [PMID: 33554861 PMCID: PMC7891932 DOI: 10.7554/elife.58838] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 02/05/2021] [Indexed: 12/22/2022] Open
Abstract
Innate lymphoid cells (ILCs) represent the most recently identified subset of effector lymphocytes, with key roles in the orchestration of early immune responses. Despite their established involvement in the pathogenesis of many inflammatory disorders, the role of ILCs in cancer remains poorly defined. Here we assessed whether human ILCs can actively interact with the endothelium to promote tumor growth control, favoring immune cell adhesion. We show that, among all ILC subsets, ILCPs elicited the strongest upregulation of adhesion molecules in endothelial cells (ECs) in vitro, mainly in a contact-dependent manner through the tumor necrosis factor receptor- and RANK-dependent engagement of the NF-κB pathway. Moreover, the ILCP-mediated activation of the ECs resulted to be functional by fostering the adhesion of other innate and adaptive immune cells. Interestingly, pre-exposure of ILCPs to human tumor cell lines strongly impaired this capacity. Hence, the ILCP-EC interaction might represent an attractive target to regulate the immune cell trafficking to tumor sites and, therefore, the establishment of an anti-tumor immune response.
Collapse
Affiliation(s)
- Giulia Vanoni
- Department of Oncology, Ludwig Institute for Cancer Research - University of LausanneLausanneSwitzerland
| | - Giuseppe Ercolano
- Department of Oncology, Ludwig Institute for Cancer Research - University of LausanneLausanneSwitzerland
| | - Simona Candiani
- Department of Earth Science, Environment and Life, University of GenovaGenovaItaly
| | - Mariangela Rutigliani
- Department of Laboratory and Service, Histological and Anatomical Pathology, E.O. Galliera HospitalGenovaItaly
| | - Mariangela Lanata
- Department of Laboratory and Service, Histological and Anatomical Pathology, E.O. Galliera HospitalGenovaItaly
| | - Laurent Derré
- Department of Urology, University Hospital of Lausanne (CHUV)LausanneSwitzerland
| | - Emanuela Marcenaro
- Department of Experimental Medicine and Centre of Excellence for Biomedical Research, University of GenovaGenovaItaly
| | - Pascal Schneider
- Department of Biochemistry, University of LausanneLausanneSwitzerland
| | - Pedro Romero
- Department of Oncology, University of LausanneLausanneSwitzerland
| | - Camilla Jandus
- Department of Oncology, Ludwig Institute for Cancer Research - University of LausanneLausanneSwitzerland
| | - Sara Trabanelli
- Department of Oncology, Ludwig Institute for Cancer Research - University of LausanneLausanneSwitzerland
| |
Collapse
|
18
|
Li Y, Yan H, Guo J, Han Y, Zhang C, Liu X, Du J, Tian XL. Down-regulated RGS5 by genetic variants impairs endothelial cell function and contributes to coronary artery disease. Cardiovasc Res 2021; 117:240-255. [PMID: 31605122 DOI: 10.1093/cvr/cvz268] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/22/2019] [Accepted: 10/04/2019] [Indexed: 12/20/2022] Open
Abstract
AIMS Genetic contribution to coronary artery disease (CAD) remains largely unillustrated. Although transcriptomic profiles have identified dozens of genes that are differentially expressed in normal and atherosclerotic vessels, whether those genes are genetically associated with CAD remains to be determined. Here, we combined genetic association studies, transcriptome profiles and in vitro and in vivo functional experiments to identify novel susceptibility genes for CAD. METHODS AND RESULTS Through an integrative analysis of transcriptome profiles with genome-wide association studies for CAD, we obtained 18 candidate genes and selected one representative single nucleotide polymorphism (SNP) for each gene for multi-centred validations. We identified an intragenic SNP, rs1056515 in RGS5 gene (odds ratio = 1.17, 95% confidence interval =1.10-1.24, P = 3.72 × 10-8) associated with CAD at genome-wide significance. Rare genetic variants in linkage disequilibrium with rs1056515 were identified in CAD patients leading to a decreased expression of RGS5. The decreased expression was also observed in atherosclerotic vessels and endothelial cells treated by various cardiovascular risk factors. Through siRNA knockdown and adenoviral overexpression, we further showed that RGS5 regulated endothelial inflammation, vascular remodelling, as well as canonical NF-κB signalling activation. Moreover, CXCL12, a specific downstream target of the non-canonical NF-κB pathway, was strongly affected by RGS5. However, the p100 processing, a well-documented marker for non-canonical NF-κB pathway activation, was not altered, suggesting an existence of a novel mechanism by which RGS5 regulates CXCL12. CONCLUSIONS We identified RGS5 as a novel susceptibility gene for CAD and showed that the decreased expression of RGS5 impaired endothelial cell function and functionally contributed to atherosclerosis through a variety of molecular mechanisms. How RGS5 regulates the expression of CXCL12 needs further studies.
Collapse
Affiliation(s)
- Yang Li
- Vascular Biology Laboratory, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
| | - Han Yan
- Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, No. 5 Yiheyuan Road, Beijing, China
| | - Jian Guo
- Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, No. 5 Yiheyuan Road, Beijing, China
| | - Yingchun Han
- Vascular Biology Laboratory, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
| | - Cuifang Zhang
- Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, No. 5 Yiheyuan Road, Beijing, China
| | - Xiuying Liu
- Center for Molecular Systems Biology, Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Jie Du
- Vascular Biology Laboratory, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
| | - Xiao-Li Tian
- Department of Human Population Genetics, Institute of Molecular Medicine, Peking University, No. 5 Yiheyuan Road, Beijing, China
- Department of Human Population Genetics, A217 Life Science Building, Human Aging Research Institute and School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, 999 Xuefu Road, Honggutan New District, Nanchang City, Jiangxi Province 330031, China
| |
Collapse
|
19
|
Li J, Tang RS, Shi Z, Li JQ. Nuclear factor‐κB in rheumatoid arthritis. Int J Rheum Dis 2020; 23:1627-1635. [PMID: 32965792 DOI: 10.1111/1756-185x.13958] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/08/2020] [Accepted: 08/09/2020] [Indexed: 02/02/2023]
Affiliation(s)
- Jie Li
- School of Medicine, University of Electronic Science and Technology of China & department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu, China
| | - Rong-Shuang Tang
- School of Medicine, University of Electronic Science and Technology of China & department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu, China
| | - Zhou Shi
- School of Medicine, University of Electronic Science and Technology of China & department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu, China
| | - Jin-Qi Li
- School of Medicine, University of Electronic Science and Technology of China & department of Pharmacy, Sichuan Provincial People's Hospital, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province & Sichuan Academy of Medical Sciences, Chengdu, China
| |
Collapse
|
20
|
Roles of miR-640 and Zinc Finger Protein 91 (ZFP91) in Angiopoietin-1-Induced In Vitro Angiogenesis. Cells 2020; 9:cells9071602. [PMID: 32630670 PMCID: PMC7408170 DOI: 10.3390/cells9071602] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022] Open
Abstract
Angiopoietin-1 (Ang-1) is a ligand of Tie-2 receptors that promotes angiogenesis. It has been established that regulatory loops exist between angiogenic growth factors and distinct pro or anti-angiogenic miRNAs, but the nature and the roles of Ang-1-regulated miRNAs remain unclear. In this study, we assessed the role of miR-640 in Ang-1-induced angiogenesis in human umbilical vein endothelial cells (HUVECs). Exposure to Ang-1 (300 ng/mL) from 6 to 72 h significantly decreased expression of mature miR-640, a response that was mediated by Tie-2 receptors and was also observed in response to Ang-2, the vascular endothelial growth factor, and transforming growth factor β. Increasing miR-640 levels using a mimic inhibited Ang-1-induced cell migration and capillary-like tube formation whereas inhibition of miR-640 enhanced these responses. Pull down assays of biotinylated miR-640 revealed that miR-640 directly targets Zinc Finger Protein 91 (ZFP91), an atypical E3-ubiquitin ligase. Ang-1 exposure induced ZFP91 expression through down-regulation of miR-640. Silencing of ZFP91 significantly inhibited Ang-1-induced cell migration and tube formation. We conclude that Ang-1 upregulates ZFP91 expression through transcriptional down-regulation of miR-640 and that ZFP91 plays important roles in the promotion of Ang-1-induced endothelial cell migration and differentiation.
Collapse
|
21
|
Gindzienska-Sieskiewicz E, Distler O, Reszec J, Jordan S, Bielecki P, Sieskiewicz A, Sulik A, Lukasik M, Bielecki M, Kowal K, Kowal-Bielecka O. Increased expression of the TNF superfamily member LIGHT/TNFSF14 and its receptors (HVEM and LTßR) in patients with systemic sclerosis. Rheumatology (Oxford) 2020; 58:502-510. [PMID: 30508197 DOI: 10.1093/rheumatology/key348] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 10/18/2018] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES This study aimed to assess the potential role of the TNF superfamily member lymphocyte T-related inducible ligand that competes for glycoprotein D binding to herpesvirus entry mediator on T cells (LIGHT) in SSc through evaluation of: skin expression of LIGHT and its receptors, herpesvirus entry mediator and lymphotoxin ß-related receptor, and serum concentration of LIGHT in SSc patients. METHODS Expression of LIGHT and its receptors was investigated by immunohistochemistry and evaluated semi-quantitatively in skin biopsies from 19 SSc patients and 9 healthy controls. Serum levels of LIGHT were measured using ELISA in 329 patients with SSc and 50 control subjects. RESULTS Expression of LIGHT and both receptors was higher in SSc patients compared with controls (P < 0.05 for all comparisons). Patients with early SSc (⩽ 3 years from the first non-Raynaud's phenomenon symptom) showed higher expression of LIGHT and herpesvirus entry mediator compared with patients with longer disease duration (P < 0.05 for both comparisons). The mean serum concentration of LIGHT was significantly higher in SSc patients compared with the controls (P < 0.05). High serum concentration of LIGHT was associated with male sex, presence of digital ulcers, muscle involvement (defined by elevated serum creatine kinase levels), steroid treatment and lack of ACA. However, in multivariate regression analysis only presence of digital ulcers and creatine kinase elevation were independently associated with serum concentration of LIGHT. CONCLUSION These data provide the first evidence of overexpression of LIGHT and its receptors in SSc and suggest that the LIGHT axis might contribute to the pathogenesis of SSc. Increased serum concentrations of LIGHT seem to reflect vascular injury in SSc.
Collapse
Affiliation(s)
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Joanna Reszec
- Department of Medical Pathomorphology, Medical University of Bialystok, Bialystok, Poland
| | - Suzana Jordan
- Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Pawel Bielecki
- Department of Otolaryngology, Medical University of Bialystok, Bialystok, Poland
| | - Andrzej Sieskiewicz
- Department of Otolaryngology, Medical University of Bialystok, Bialystok, Poland
| | - Agnieszka Sulik
- Department of Rheumatology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Malgorzata Lukasik
- Department of Medical Pathomorphology, Medical University of Bialystok, Bialystok, Poland
| | - Marek Bielecki
- Department of Orthopedics and Traumatology, Medical University of Bialystok, Bialystok, Poland
| | - Krzysztof Kowal
- Department of Allergology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland.,Department of Experimental Allergology and Immunology, Medical University of Bialystok, Bialystok, Poland
| | - Otylia Kowal-Bielecka
- Department of Rheumatology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
22
|
A potential role of toll-like receptors, IFN-γ and the phosphatidylinositol 3-kinase pathway in the pathogenesis of acquired mediastinal lymphatic malformation. Med Hypotheses 2019; 131:109287. [PMID: 31443764 DOI: 10.1016/j.mehy.2019.109287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/23/2019] [Indexed: 11/20/2022]
Abstract
Sarcoidosis is a multisystem disorder with non-caseating granulomas in various organs. The etiology of sarcoid granuloma formation is not clear and likely an antigen-induced process. We came across a previously treated sarcoidosis patient who presented with worsening dyspnea on exertion for several months and several days of difficulty swallowing. On Chest CT imaging, large posterior mediastinal mass was found that subsequently diagnosed as macrocystic lymphatic malformation after surgical resection. Pathophysiology of development of acquired lymphatic malformations in a sarcoidosis patient is currently not clear. We hypothesize there might be a complex interplay of Toll-like receptors, IFN-γ and the phosphatidylinositol 3-kinase pathway in the pathogenesis.
Collapse
|
23
|
Xie CB, Qin L, Li G, Fang C, Kirkiles-Smith NC, Tellides G, Pober JS, Jane-Wit D. Complement Membrane Attack Complexes Assemble NLRP3 Inflammasomes Triggering IL-1 Activation of IFN-γ-Primed Human Endothelium. Circ Res 2019; 124:1747-1759. [PMID: 31170059 DOI: 10.1161/circresaha.119.314845] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
RATIONALE Complement activation contributes to multiple immune-mediated pathologies. In late allograft failure, donor-specific antibody deposits complement membrane attack complexes (MAC) on graft endothelial cells (ECs), substantially increasing their immunogenicity without causing lysis. Internalized MAC stabilize NIK (NF-κB [nuclear factor kappa-light-chain-enhancer of activated B cells]-inducing kinase) protein on Rab5+MAC+ endosomes, activating noncanonical NF-κB signaling. However, the link to increased immunogenicity is unclear. OBJECTIVE To identify mechanisms by which alloantibody and internalized MAC activate ECs to enhance their ability to increase T-cell responses. METHODS AND RESULTS In human EC cultures, internalized MAC also causes NLRP3 (NOD-like receptor family pyrin domain containing 3) translocation from endoplasmic reticulum to Rab5+MAC+NIK+ endosomes followed by endosomal NIK-dependent inflammasome assembly. Cytosolic NIK, stabilized by LIGHT (lymphotoxin-like inducible protein that competes with glycoprotein D for herpesvirus entry on T cells), does not trigger inflammasome assembly, and ATP-triggered inflammasome assembly does not require NIK. IFN-γ (interferon-γ) primes EC responsiveness to MAC by increasing NLRP3, pro-caspase 1, and gasdermin D expression. NIK-activated noncanonical NF-κB signaling induces pro-IL (interleukin)-1β expression. Inflammasome processed pro-IL-1β, and gasdermin D results in IL-1β secretion that increases EC immunogenicity through IL-1 receptor signaling. Activation of human ECs lining human coronary artery grafts in immunodeficient mouse hosts by alloantibody and complement similarly depends on assembly of an NLRP3 inflammasome. Finally, in renal allograft biopsies showing chronic rejection, caspase-1 is activated in C4d+ ECs of interstitial microvessels, supporting the relevance of the cell culture findings. CONCLUSIONS In response to antibody-mediated complement activation, IFN-γ-primed human ECs internalize MAC, triggering both endosomal-associated NIK-dependent NLRP3 inflammasome assembly and IL-1 synthesis, resulting in autocrine/paracrine IL-1β-mediated increases in EC immunogenicity. Similar responses may underlie other complement-mediated pathologies.
Collapse
Affiliation(s)
- Catherine B Xie
- From the Department of Immunobiology (C.B.X., N.C.K.-S., J.S.P.), Yale University School of Medicine, New Haven, CT
| | - Lingfeng Qin
- Department of Surgery (L.Q., G.L., G.T.), Yale University School of Medicine, New Haven, CT
| | - Guangxin Li
- Department of Surgery (L.Q., G.L., G.T.), Yale University School of Medicine, New Haven, CT
| | - Caodi Fang
- Division of Cardiovascular Medicine (C.F., D.J.-w), Yale University School of Medicine, New Haven, CT
| | - Nancy C Kirkiles-Smith
- From the Department of Immunobiology (C.B.X., N.C.K.-S., J.S.P.), Yale University School of Medicine, New Haven, CT
| | - George Tellides
- Department of Surgery (L.Q., G.L., G.T.), Yale University School of Medicine, New Haven, CT
| | - Jordan S Pober
- From the Department of Immunobiology (C.B.X., N.C.K.-S., J.S.P.), Yale University School of Medicine, New Haven, CT
| | - Dan Jane-Wit
- Division of Cardiovascular Medicine (C.F., D.J.-w), Yale University School of Medicine, New Haven, CT
| |
Collapse
|
24
|
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
|
25
|
Jiang T, Kai D, Liu S, Huang X, Heng S, Zhao J, Chan BQY, Loh XJ, Zhu Y, Mao C, Zheng L. Mechanically cartilage-mimicking poly(PCL-PTHF urethane)/collagen nanofibers induce chondrogenesis by blocking NF-kappa B signaling pathway. Biomaterials 2018; 178:281-292. [PMID: 29945065 PMCID: PMC6301151 DOI: 10.1016/j.biomaterials.2018.06.023] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/26/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022]
Abstract
Cartilage cannot self-repair and thus regeneration is a promising approach to its repair. Here we developed new electrospun nanofibers, made of poly (ε-caprolactone)/polytetrahydrofuran (PCL-PTHF urethane) and collagen I from calf skin (termed PC), to trigger the chondrogenic differentiation of mesenchymal stem cells (MSCs) and the cartilage regeneration in vivo. We found that the PC nanofibers had a modulus (4.3 Mpa) lower than the PCL-PTHF urethane nanofibers without collagen I from calf skin (termed P) (6.8 Mpa) although both values are within the range of the modulus of natural cartilage (1-10 MPa). Both P and PC nanofibers did not show obvious difference in the morphology and size. Surprisingly, in the absence of the additional chondrogenesis inducers, the softer PC nanofibers could induce the chondrogenic differentiation in vitro and cartilage regeneration in vivo more efficiently than the stiffer P nanofibers. Using mRNA-sequence analysis, we found that the PC nanofibers outperformed P nanofibers in inducing chondrogenesis by specifically blocking the NF-kappa B signaling pathway to suppress inflammation. Our work shows that the PC nanofibers can serve as building blocks of new scaffolds for cartilage regeneration and provides new insights on the effect of the mechanical properties of the nanofibers on the cartilage regeneration.
Collapse
Affiliation(s)
- Tongmeng Jiang
- Department of Bone and Joint Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China; Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Dan Kai
- Institute of Materials Research and Engineering (IMRE), A*STAR, 3 Research Link, Singapore 117602, Singapore
| | - Sijia Liu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China; School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, China
| | - Xianyuan Huang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Shujun Heng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Jinmin Zhao
- Department of Bone and Joint Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China; Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China.
| | - Benjamin Qi Yu Chan
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), A*STAR, 3 Research Link, Singapore 117602, Singapore; Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore; Singapore Eye Research Institute, 11 Third Hospital Avenue, Singapore 168751, Singapore
| | - Ye Zhu
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019-5300, USA
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019-5300, USA; School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
26
|
D'Ignazio L, Batie M, Rocha S. TNFSF14/LIGHT, a Non-Canonical NF-κB Stimulus, Induces the HIF Pathway. Cells 2018; 7:E102. [PMID: 30096845 PMCID: PMC6116154 DOI: 10.3390/cells7080102] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/06/2018] [Accepted: 08/07/2018] [Indexed: 12/20/2022] Open
Abstract
Non-canonical NF-κB signalling plays important roles in the development and function of the immune system but it also is deregulated in a number of inflammatory diseases. Although, NF-κB and HIF crosstalk has been documented, this has only been described following canonical NF-κB stimulation, involving RelA/p50 and the HIF-1 dimer. Here, we report that the non-canonical inducer TNFSF14/LIGHT leads to HIF induction and activation in cancer cells. We demonstrate that only HIF-2α is induced at the transcriptional level following non-canonical NF-κB activation, via a mechanism that is dependent on the p52 subunit. Furthermore, we demonstrate that p52 can bind to the HIF-2α promoter in cells. These results indicate that non-canonical NF-κB can lead to HIF signalling implicating HIF-2α as one of the downstream effectors of this pathway in cells.
Collapse
Affiliation(s)
- Laura D'Ignazio
- Center for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD15EH, UK.
| | - Michael Batie
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L697ZB, UK.
| | - Sonia Rocha
- Center for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD15EH, UK.
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L697ZB, UK.
| |
Collapse
|
27
|
Piao W, Xiong Y, Famulski K, Brinkman CC, Li L, Toney N, Wagner C, Saxena V, Simon T, Bromberg JS. Regulation of T cell afferent lymphatic migration by targeting LTβR-mediated non-classical NFκB signaling. Nat Commun 2018; 9:3020. [PMID: 30069025 PMCID: PMC6070541 DOI: 10.1038/s41467-018-05412-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 06/02/2018] [Indexed: 12/23/2022] Open
Abstract
Lymphotoxin-beta receptor (LTβR) signaling in lymphatic endothelial cells (LEC) regulates leukocyte afferent lymphatic transendothelial migration (TEM). The function of individual signaling pathways for different leukocyte subsets is currently unknown. Here, we show that LTβR signals predominantly via the constitutive and ligand-driven non-classical NIK pathway. Targeting LTβR-NIK by an LTβR-derived decoy peptide (nciLT) suppresses the production of chemokines CCL21 and CXCL12, and enhances the expression of classical NFκB-driven VCAM-1 and integrin β4 to retain T cells on LEC and precludes T cell and dendritic cell TEM. nciLT inhibits contact hypersensitivity (CHS) at both the sensitization and elicitation stages, likely by inhibiting leukocyte migration. By contrast, targeting LTβR-classical NFκB signaling during the elicitation and resolution stages attenuates CHS, possibly by promoting leukocyte egress. These findings demonstrate the importance of LTβR signaling in leukocyte migration and LEC and lymphatic vessel function, and show that antagonist peptides may serve as lead compounds for therapeutic applications.
Collapse
Affiliation(s)
- Wenji Piao
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yanbao Xiong
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Konrad Famulski
- Department of Laboratory Medicine and Pathology, University of Alberta, 250 Heritage Medical Research Centre, Edmonton, AB, T6G 2S2, Canada
| | - C Colin Brinkman
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Lushen Li
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Nicholas Toney
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Chelsea Wagner
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Vikas Saxena
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Thomas Simon
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jonathan S Bromberg
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
28
|
MacroH2A1.2 inhibits prostate cancer-induced osteoclastogenesis through cooperation with HP1α and H1.2. Oncogene 2018; 37:5749-5765. [PMID: 29925860 PMCID: PMC6309402 DOI: 10.1038/s41388-018-0356-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 05/13/2018] [Accepted: 05/18/2018] [Indexed: 12/22/2022]
Abstract
Osteoclasts are multinuclear bone-resorbing cells that differentiate from hematopoietic precursor cells. Prostate cancer cells frequently spread to bone and secrete soluble signaling factors to accelerate osteoclast differentiation and bone resorption. However, processes and mechanisms that govern the expression of osteoclastogenic soluble factors secreted by prostate cancer cells are largely unknown. MacroH2A (mH2A) is a histone variant that replaces canonical H2A at designated genomic loci and establishes functionally distinct chromatin regions. Here we report that mH2A1.2, one of the mH2A isoforms, attenuates prostate cancer-induced osteoclastogenesis by maintaining the inactive state of genes encoding soluble factors in prostate cancer cells. Our functional analyses of soluble factors identify lymphotoxin beta (LTβ) as a major stimulator of osteoclastogenesis and an essential mH2A1.2 target for its anti-osteoclastogenic activity. Mechanistically, mH2A1.2 directly interacts with HP1α and H1.2 and requires them to inactivate LTβ gene in prostate cancer cells. Consistently, HP1α and H1.2 have an intrinsic ability to inhibit osteoclast differentiation in a mH2A1.2-dependent manner. Together, our data uncover a new and specific role for mH2A1.2 in modulating osteoclastogenic potential of prostate cancer cells and demonstrate how this signaling pathway can be exploited to treat osteolytic bone metastases at the molecular level.
Collapse
|
29
|
Yin C, Cai XB, Wang HJ, Gu BJ, Yang XF, Zhang R, Ji XH. Pathological significance and regulatory mechanism of lymphotoxin β receptor overexpression in T cells of patients with systemic lupus erythematosus. J Biomed Res 2018; 32:113-122. [PMID: 28963441 PMCID: PMC5895565 DOI: 10.7555/jbr.27.20130046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a typical autoimmune disease. Lymphotoxin β receptor (LTβR) signaling plays an important role in autoimmune inflammations. LTβR-Ig fusion protein, LTβR blocking agent, has been used to treat SLE, while its mechanism remains to be fully elucidated. In this study, to investigate the expression of LTβR in the T cells of SLE patients and its roles in the pathogenesis of SLE, we isolated the peripheral blood T cells of SLE patients and normal controls to detect expression of LTβR by flow cytometry and RNA assay. T cells were also stimulated with LIGHT, a ligand of LTβR, and then detected for their LTβR expressions and apoptosis by flow cytometry. Also, their expressions of inflammatory factors and receptors were determined by RNA assay. The results showed that LTβR positive cells were 22.75%±6.98% in CD3+ cells of SLE patients, while there were almost no LTβR positive cells in CD3+ cells of normal persons. Moreover, LTβR expression was remarkably higher in CD3, CD4 and CD8 positive T cells of active SLE patients than non/low active patients (all P<0.05), and positively correlated with increased Ig level, decreased complement level and renal damage. Moreover, the stimulation of SLE T cells with LIGHT promoted higher expression of LTβR, IL-23R and IL-17A, and apoptosis of T cells. In conclusion, we demonstrated a high expression of LTβR in the T cells of SLE patients which may be associated with pathogenesis of SLE.
Collapse
Affiliation(s)
- Cheng Yin
- Department of Immunology, Basic Medical School, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Red Cross Blood Center, Nanjing, Jiangsu 210003, China
| | - Xu-Bing Cai
- Red Cross Blood Center, Nanjing, Jiangsu 210003, China
| | - Hui-Juan Wang
- Department of Immunology, Basic Medical School, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bing-Jie Gu
- Rheumatology Department of Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, China
| | - Xiao-Fan Yang
- Department of Immunology, Basic Medical School, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Rong Zhang
- Department of Immunology, Basic Medical School, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiao-Hui Ji
- Department of Immunology, Basic Medical School, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
30
|
Maracle CX, Agca R, Helder B, Meeuwsen JAL, Niessen HWM, Biessen EAL, de Winther MPJ, de Jager SCA, Nurmohamed MT, Tas SW. Noncanonical NF-κB signaling in microvessels of atherosclerotic lesions is associated with inflammation, atheromatous plaque morphology and myocardial infarction. Atherosclerosis 2018; 270:33-41. [PMID: 29407886 DOI: 10.1016/j.atherosclerosis.2018.01.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 12/31/2017] [Accepted: 01/18/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND AIMS Neovascularization is associated with atherosclerotic plaque instability and increased chance of myocardial infarction (MI). Patients with chronic inflammatory diseases (CID) have increased risk of atherosclerosis, and evidence demonstrates that NF-κB inducing kinase (NIK)-mediated noncanonical NF-κB signaling in endothelial cells (EC) is linked to inflammation and angiogenesis. Here, we hypothesized NIK may also be activated in EC of atherosclerotic lesion microvessels. METHODS Using cohorts of atherosclerotic lesions from coronary and carotid arteries, we quantified NIK expression in plaque microvessels and compared it to pathological markers, including inflammatory cell content, plaque characteristics and MI. Differences in gene transcripts were evaluated between stable and ruptured lesions. RESULTS NIK+EC were present in both coronary and carotid lesions. In CID patients, plaques with stenosis >40% had an increased number of NIK+EC and higher content of immune cells (p < .05) as compared to controls. Immune cells per NIK+EC were also greater in CID patients (p < .05), with pronounced differences as stenosis increased. In unstable lesions, NIK+EC were elevated as were EC expressing CXCL12 (p < .05). NIK+EC were increased in lesions with lipid content >40% (p < .05) and more abundant in coronary artery lesions implicated in MI (p < .05). These vessels also associated with atheromatous rather than fibrous plaque morphology (p < .05). Transcriptomic profiling demonstrated components of noncanonical NF-κB pathway were also upregulated in ruptured plaques (p < .05). CONCLUSIONS NIK+EC associate with chronic inflammation in advanced lesions and are linked to markers of local inflammation, lipid content, unstable plaque phenotype and development of MI. Therefore, targeting noncanonical NF-κB signaling may hold therapeutic potential for patients with atherosclerosis and cardiovascular disease.
Collapse
Affiliation(s)
- Chrissta X Maracle
- Amsterdam Rheumatology and Immunology Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Rabia Agca
- Amsterdam Rheumatology and Immunology Center, READE, Amsterdam, The Netherlands; Amsterdam Rheumatology and Immunology Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Boy Helder
- Amsterdam Rheumatology and Immunology Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - John A L Meeuwsen
- Laboratory for Experimental Cardiology, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Hans W M Niessen
- Amsterdam Rheumatology and Immunology Center, READE, Amsterdam, The Netherlands; Amsterdam Rheumatology and Immunology Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Erik A L Biessen
- Department of Experimental Vascular Pathology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Menno P J de Winther
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Saskia C A de Jager
- Laboratory for Experimental Cardiology, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Mike T Nurmohamed
- Amsterdam Rheumatology and Immunology Center, READE, Amsterdam, The Netherlands; Amsterdam Rheumatology and Immunology Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
31
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
32
|
Seigner J, Basilio J, Resch U, de Martin R. CD40L and TNF both activate the classical NF-κB pathway, which is not required for the CD40L induced alternative pathway in endothelial cells. Biochem Biophys Res Commun 2017; 495:1389-1394. [PMID: 29183724 DOI: 10.1016/j.bbrc.2017.11.160] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 11/24/2017] [Indexed: 11/16/2022]
Abstract
CD40L and TNF signal through engagement of their respective receptors, which are both members of the TNF receptor family. They use partially common signaling molecules leading, among others, to activation of the NF-κB pathway. However, whereas TNF activates the classical, CD40L has been reported to activate the alternative NF-κB pathway, leading to the anticipation that differences in the pattern of inflammatory gene expression would occur. Here, we have compared the gene expression repertoire of CD40L (CD154) and TNF stimulated HUVEC and report that unexpectedly, apart from a stronger response to TNF, no major qualitative differences could be observed. This applies for the period of up to 6 h, a time where the alternative pathway has already been activated. Analysis of the early events after receptor engagement revealed that both TNF and CD40L activate the classical NF-κB pathway, and confirm activation of the alternative by the latter. Furthermore, using genetic and pharmacological inhibition of the classical pathway we show that activation of the alternative occurs independently of the former. This reveals novel insights into NF-κB signaling by CD40L and TNF in endothelial cells.
Collapse
Affiliation(s)
- J Seigner
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - J Basilio
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - U Resch
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - R de Martin
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, A-1090 Vienna, Austria.
| |
Collapse
|
33
|
Yang JG, Sun YF, He KF, Ren JG, Liu ZJ, Liu B, Zhang W, Zhao YF. Lymphotoxins Promote the Progression of Human Lymphatic Malformation by Enhancing Lymphatic Endothelial Cell Proliferation. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2602-2615. [DOI: 10.1016/j.ajpath.2017.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/24/2017] [Accepted: 07/26/2017] [Indexed: 12/31/2022]
|
34
|
Pursani V, Pethe P, Bashir M, Sampath P, Tanavde V, Bhartiya D. Genetic and Epigenetic Profiling Reveals EZH2-mediated Down Regulation of OCT-4 Involves NR2F2 during Cardiac Differentiation of Human Embryonic Stem Cells. Sci Rep 2017; 7:13051. [PMID: 29026152 PMCID: PMC5638931 DOI: 10.1038/s41598-017-13442-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/25/2017] [Indexed: 02/07/2023] Open
Abstract
Human embryonic (hES) stem cells are widely used as an in vitro model to understand global genetic and epigenetic changes that occur during early embryonic development. In-house derived hES cells (KIND1) were subjected to directed differentiation into cardiovascular progenitors (D12) and beating cardiomyocytes (D20). Transcriptome profiling of undifferentiated (D0) and differentiated (D12 and 20) cells was undertaken by microarray analysis. ChIP and sequential ChIP were employed to study role of transcription factor NR2F2 during hES cells differentiation. Microarray profiling showed that an alteration of about 1400 and 1900 transcripts occurred on D12 and D20 respectively compared to D0 whereas only 19 genes were altered between D12 and D20. This was found associated with corresponding expression pattern of chromatin remodelers, histone modifiers, miRNAs and lncRNAs marking the formation of progenitors and cardiomyocytes on D12 and D20 respectively. ChIP sequencing and sequential ChIP revealed the binding of NR2F2 with polycomb group member EZH2 and pluripotent factor OCT4 indicating its crucial involvement in cardiac differentiation. The study provides a detailed insight into genetic and epigenetic changes associated with hES cells differentiation into cardiac cells and a role for NR2F2 is deciphered for the first time to down-regulate OCT-4 via EZH2 during cardiac differentiation.
Collapse
Affiliation(s)
- Varsha Pursani
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive Health, Mumbai, 400012, India
| | - Prasad Pethe
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive Health, Mumbai, 400012, India
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS University, Mumbai, 400056, India
| | - Mohsin Bashir
- Institute of Medical Biology, Agency for Science Technology & Research (A*STAR), Singapore, 138648, Singapore
| | - Prabha Sampath
- Institute of Medical Biology, Agency for Science Technology & Research (A*STAR), Singapore, 138648, Singapore
| | - Vivek Tanavde
- Bioinformatics Institute, Agency for Science Technology & Research (A*STAR), Singapore, 138671, Singapore
- Division of Biological & Life Sciences, School of Arts & Sciences, Ahmedabad University, Ahmedabad, 380009, India
| | - Deepa Bhartiya
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive Health, Mumbai, 400012, India.
| |
Collapse
|
35
|
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: 99] [Impact Index Per Article: 12.4] [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
|
36
|
Maracle CX, Kucharzewska P, Helder B, van der Horst C, Correa de Sampaio P, Noort AR, van Zoest K, Griffioen AW, Olsson H, Tas SW. Targeting non-canonical nuclear factor-κB signalling attenuates neovascularization in a novel 3D model of rheumatoid arthritis synovial angiogenesis. Rheumatology (Oxford) 2017; 56:294-302. [PMID: 27864565 DOI: 10.1093/rheumatology/kew393] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 10/03/2016] [Indexed: 12/16/2023] Open
Abstract
OBJECTIVE Angiogenesis is crucial in RA disease progression. Lymphotoxin β receptor (LTβR)-induced activation of the non-canonical nuclear factor-κB (NF-κB) pathway via NF-κB-inducing kinase (NIK) has been implicated in this process. Consequently, inhibition of this pathway may hold therapeutic potential in RA. We describe a novel three-dimensional (3D) model of synovial angiogenesis incorporating endothelial cells (ECs), RA fibroblast-like synoviocytes (RAFLSs) and RA synovial fluid (RASF) to further investigate the contributions of NF-κB in this process. METHODS Spheroids consisting of RAFLSs and ECs were stimulated with RASF, the LTβR ligands LTβ and LIGHT, or growth factor bFGF and VEGF, followed by quantification of EC sprouting using confocal microscopy and digital image analysis. Next, the effects of anginex, NIK-targeting siRNA (siNIK), LTβR-Ig fusion protein (baminercept) and a novel pharmacological NIK inhibitor were investigated. RESULTS RASF significantly promoted sprout formation, which was blocked by the established angiogenesis inhibitor anginex (P < 0.05). LTβ and LIGHT induced significant sprouting (P < 0.05), as did bFGF/VEGF (P < 0.01). siNIK pre-treatment of ECs led to reductions in LTβR-induced vessel formation (P < 0.05). LTβR-Ig not only blocked LTβ- or LIGHT-induced sprouting, but also RASF-induced sprouting (P < 0.05). The NIK inhibitor blocked angiogenesis induced by LTβ, LIGHT, growth factors (P < 0.05) and RASF (P < 0.01). CONCLUSION We present a novel 3D model of synovial angiogenesis incorporating RAFLSs, ECs and RASF that mimics the in vivo situation. Using this system, we demonstrate that non-canonical NF-κB signalling promotes neovascularization and show that this model is useful for dissecting relative contributions of signalling pathways in specific cell types to angiogenic responses and for testing pharmacological inhibitors of angiogenesis.
Collapse
Affiliation(s)
- Chrissta X Maracle
- Amsterdam Rheumatology and immunology Center
- Department of Clinical Immunology & Rheumatology
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Boy Helder
- Amsterdam Rheumatology and immunology Center
- Department of Clinical Immunology & Rheumatology
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Corine van der Horst
- Amsterdam Rheumatology and immunology Center
- Department of Clinical Immunology & Rheumatology
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Ae-Ri Noort
- Amsterdam Rheumatology and immunology Center
- Department of Clinical Immunology & Rheumatology
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Katinka van Zoest
- Amsterdam Rheumatology and immunology Center
- Department of Clinical Immunology & Rheumatology
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Sander W Tas
- Amsterdam Rheumatology and immunology Center
- Department of Clinical Immunology & Rheumatology
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Wang M, Wang L, Huang C, Wang IW, Turrentine MW. Regulation of myocardial stromal cell-derived factor 1α/CXCL12 by tumor necrosis factor signaling. J Surg Res 2016; 207:155-163. [PMID: 27979472 DOI: 10.1016/j.jss.2016.08.073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 08/05/2016] [Accepted: 08/24/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Global myocardial ischemia-reperfusion (I/R) occurs during cardiac operations. This I/R injury leads to increased production of tumor necrosis factor α (TNF) instantly and upregulated expression of stromal cell-derived factor 1 α (SDF-1). On the basis of the published data from our laboratory and other groups, locally produced TNF contributes to cardiac dysfunction mainly via binding to its receptor (tumor necrosis factor receptor 1 [TNFR1]), whereas ischemia-induced myocardial SDF-1 mediates cardioprotection. Although TNF has been shown to work as an upstream initiator for induction of other cytokines and chemokines, there is no information regarding the interaction among TNF, TNFRs, and myocardial SDF-1 expression. In this study, given that TNF downregulated SDF-1 in vascular endothelial cells, we therefore hypothesized that TNF would have a negative effect on myocardial SDF-1 production, which is attributable to TNFR-initiated actions. METHODS Using a Langendorff model, isolated male mouse hearts were infused with TNF for 45 min. Male adult mouse hearts from wild type, TNFR1 knockout (TNFR1KO), TNFR2KO, and TNFR1/2KO were subjected to global I/R. H9c2 cells with small interfering RNA transfection were used as an in vitro model. The levels of SDF-1 (protein and messenger RNA) were detected by enzyme-linked immunosorbent assay and quantitative reverse transcription-polymerase chain reaction . Protein kinases of IκB (nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor α) and c-jun N-terminal kinase were also determined using Western blot assay. RESULTS TNF infusion downregulated myocardial SDF-1 production in a dose-dependent manner in the hearts. In addition, using TNF significantly decreased SDF-1 expression in cardiomyoblasts (H9c2 cells), which was associated with reduced IκB level. Knockdown of TNFR1 or TNFR2 by small interfering RNAs neutralized TNF-suppressed SDF-1 in H9c2 cells. Furthermore, deletion of TNFR1/2 or TNFR2 increased SDF-1 production in the hearts after I/R. CONCLUSIONS Our study represents the initial evidence showing that TNF plays an inhibitory role in modulating myocardial SDF-1 production and blockade of TNF signaling by ablation of TNFR1 and TNFR2 genes increased SDF-1 expression in the heart. These data expand on TNF signaling-initiated mechanisms in myocardium, which may lend a more complete understanding of SDF-1 and TNFR-derived actions in hopes of advancing ischemic heart injury treatments.
Collapse
Affiliation(s)
- Meijing Wang
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana.
| | - Lina Wang
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Chunyan Huang
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - I-Wen Wang
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mark W Turrentine
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
38
|
Treg engage lymphotoxin beta receptor for afferent lymphatic transendothelial migration. Nat Commun 2016; 7:12021. [PMID: 27323847 PMCID: PMC4919545 DOI: 10.1038/ncomms12021] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 05/20/2016] [Indexed: 02/07/2023] Open
Abstract
Regulatory T cells (Tregs) are essential to suppress unwanted immunity or inflammation. After islet allo-transplant Tregs must migrate from blood to allograft, then via afferent lymphatics to draining LN to protect allografts. Here we show that Tregs but not non-Treg T cells use lymphotoxin (LT) during migration from allograft to draining LN, and that LT deficiency or blockade prevents normal migration and allograft protection. Treg LTαβ rapidly modulates cytoskeletal and membrane structure of lymphatic endothelial cells; dependent on VCAM-1 and non-canonical NFκB signalling via LTβR. These results demonstrate a form of T-cell migration used only by Treg in tissues that serves an important role in their suppressive function and is a unique therapeutic focus for modulating suppression.
Collapse
|
39
|
Fernandes MT, Dejardin E, dos Santos NR. Context-dependent roles for lymphotoxin-β receptor signaling in cancer development. Biochim Biophys Acta Rev Cancer 2016; 1865:204-19. [PMID: 26923876 DOI: 10.1016/j.bbcan.2016.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 02/03/2016] [Accepted: 02/24/2016] [Indexed: 12/20/2022]
Abstract
The LTα1β2 and LIGHT TNF superfamily cytokines exert pleiotropic physiological functions through the activation of their cognate lymphotoxin-β receptor (LTβR). Interestingly, since the discovery of these proteins, accumulating evidence has pinpointed a role for LTβR signaling in carcinogenesis. Early studies have shown a potential anti-tumoral role in a subset of solid cancers either by triggering apoptosis in malignant cells or by eliciting an anti-tumor immune response. However, more recent studies provided robust evidence that LTβR signaling is also involved in diverse cell-intrinsic and microenvironment-dependent pro-oncogenic mechanisms, affecting several solid and hematological malignancies. Consequently, the usefulness of LTβR signaling axis blockade has been investigated as a potential therapeutic approach for cancer. Considering the seemingly opposite roles of LTβR signaling in diverse cancer types and their key implications for therapy, we here extensively review the different mechanisms by which LTβR activation affects carcinogenesis, focusing on the diverse contexts and different models assessed.
Collapse
Affiliation(s)
- Mónica T Fernandes
- Centre for Biomedical Research (CBMR), University of Algarve, Faro 8005-139, Portugal; PhD Program in Biomedical Sciences, Department of Biomedical Sciences and Medicine, University of Algarve, Faro 8005-139, Portugal
| | - Emmanuel Dejardin
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Research, Molecular Biology of Diseases, University of Liège, Liège 4000, Belgium
| | - Nuno R dos Santos
- Centre for Biomedical Research (CBMR), University of Algarve, Faro 8005-139, Portugal; Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto 4200, Portugal; Institute of Pathology and Molecular Immunology, University of Porto (IPATIMUP), Porto 4200, Portugal.
| |
Collapse
|
40
|
Barroso M, Kao D, Blom HJ, Tavares de Almeida I, Castro R, Loscalzo J, Handy DE. S-adenosylhomocysteine induces inflammation through NFkB: A possible role for EZH2 in endothelial cell activation. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:82-92. [PMID: 26506125 PMCID: PMC4674364 DOI: 10.1016/j.bbadis.2015.10.019] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 09/29/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023]
Abstract
S-adenosylhomocysteine (SAH) can induce endothelial dysfunction and activation, contributing to atherogenesis; however, its role in the activation of the inflammatory mediator NFkB has not been explored. Our aim was to determine the role of NFkB in SAH-induced activation of endothelial cells. Furthermore, we examined whether SAH, as a potent inhibitor of S-adenosylmethionine-dependent methyltransferases, suppresses the function of EZH2 methyltransferase to contribute to SAH-induced endothelial cell activation. We found that excess SAH increases the expression of adhesion molecules and cytokines in human coronary artery endothelial cells. Importantly, this up-regulation was suppressed in cells expressing a dominant negative form of the NFkB inhibitor, IkB. Moreover, SAH accumulation triggers the activation of both the canonical and non-canonical NFkB pathways, decreases EZH2, and reduces histone 3 lysine 27 trimethylation. EZH2 knockdown recapitulated the effects of excess SAH on endothelial activation, i.e., it induced NFkB activation and the subsequent up-regulation of adhesion molecules and cytokines. Our findings suggest that suppression of the epigenetic regulator EZH2 by excess SAH may contribute to NFkB activation and the consequent vascular inflammatory response. These studies unveil new targets of SAH regulation, demonstrating that EZH2 suppression and NFkB activation mediated by SAH accumulation may contribute to its adverse effects in the vasculature.
Collapse
Affiliation(s)
- Madalena Barroso
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Derrick Kao
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Henk J Blom
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, University Medical Centre Freiburg, Freiburg, Germany
| | - Isabel Tavares de Almeida
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Rita Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal; Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Joseph Loscalzo
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Diane E Handy
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Elshabrawy HA, Chen Z, Volin MV, Ravella S, Virupannavar S, Shahrara S. The pathogenic role of angiogenesis in rheumatoid arthritis. Angiogenesis 2015; 18:433-48. [PMID: 26198292 PMCID: PMC4879881 DOI: 10.1007/s10456-015-9477-2] [Citation(s) in RCA: 362] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/14/2015] [Indexed: 12/31/2022]
Abstract
Angiogenesis is the formation of new capillaries from pre-existing vasculature, which plays a critical role in the pathogenesis of several inflammatory autoimmune diseases such as rheumatoid arthritis (RA), spondyloarthropathies, psoriasis, systemic lupus erythematosus, systemic sclerosis, and atherosclerosis. In RA, excessive migration of circulating leukocytes into the inflamed joint necessitates formation of new blood vessels to provide nutrients and oxygen to the hypertrophic joint. The dominance of the pro-angiogenic factors over the endogenous angiostatic mediators triggers angiogenesis. In this review article, we highlight the underlying mechanisms by which cells present in the RA synovial tissue are modulated to secrete pro-angiogenic factors. We focus on the significance of pro-angiogenic factors such as growth factors, hypoxia-inducible factors, cytokines, chemokines, matrix metalloproteinases, and adhesion molecules on RA pathogenesis. As pro-angiogenic factors are primarily produced from RA synovial tissue macrophages and fibroblasts, we emphasize the key role of RA synovial tissue lining layer in maintaining synovitis through neovascularization. Lastly, we summarize the specific approaches utilized to target angiogenesis. We conclude that the formation of new blood vessels plays an indispensable role in RA progression. However, since the function of several pro-angiogenic mediators is cross regulated, discovering novel approaches to target multiple cascades or selecting an upstream cascade that impairs the activity of a number of pro-angiogenic factors may provide a promising strategy for RA therapy.
Collapse
Affiliation(s)
- Hatem A Elshabrawy
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, MSB 835 S Wolcott Ave., E807-E809, Chicago, IL, 60612, USA
| | - Zhenlong Chen
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, MSB 835 S Wolcott Ave., E807-E809, Chicago, IL, 60612, USA
| | - Michael V Volin
- Department of Microbiology and Immunology, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL, 60515, USA
| | - Shalini Ravella
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, MSB 835 S Wolcott Ave., E807-E809, Chicago, IL, 60612, USA
| | - Shanti Virupannavar
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, MSB 835 S Wolcott Ave., E807-E809, Chicago, IL, 60612, USA
| | - Shiva Shahrara
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, MSB 835 S Wolcott Ave., E807-E809, Chicago, IL, 60612, USA.
| |
Collapse
|
42
|
Maijer KI, Noort AR, de Hair MJH, van der Leij C, van Zoest KPM, Choi IY, Gerlag DM, Maas M, Tak PP, Tas SW. Nuclear Factor-κB-inducing Kinase Is Expressed in Synovial Endothelial Cells in Patients with Early Arthritis and Correlates with Markers of Inflammation: A Prospective Cohort Study. J Rheumatol 2015; 42:1573-81. [PMID: 26178280 DOI: 10.3899/jrheum.150245] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2015] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The nuclear factor-κB (NF-κB) family of transcription factors is strongly involved in synovial inflammation. We have previously shown that NF-κB-inducing kinase (NIK) is a key regulator of inflammation-induced angiogenesis in rheumatoid arthritis (RA) synovial tissue (ST). Here, we investigated synovial NIK expression in patients with early arthritis and in autoantibody-positive individuals at risk of developing RA. METHODS ST biopsies were obtained by arthroscopy from 154 patients with early arthritis (duration < 1 yr) with various diagnoses and 54 IgM rheumatoid factor-positive and/or anticitrullinated protein antibodies-positive individuals without evidence of arthritis. ST was stained for NIK and endothelial cell (EC) markers. Additionally, measures of disease activity were collected and contrast-enhanced magnetic resonance imaging (MRI) was performed in a subset of these patients. RESULTS In patients with early arthritis, NIK was predominantly expressed in EC of small blood vessels. Further, NIK expression correlated with erythrocyte sedimentation rate (r 0.184, p = 0.024), C-reactive protein (r 0.194, p = 0.017), joint swelling (r 0.297, p < 0.001), synovial immune cell markers (lining r 0.585, p < 0.001; sublining macrophages r 0.728, p < 0.001; T cells r 0.733, p < 0.001; and B cells r 0.264, p = 0.040), MRI effusion (r 0.665, p < 0.001), MRI synovitis (r 0.632, p < 0.001), and MRI total score (r 0.569, p < 0.001). In 18.5% of autoantibody-positive individuals, ST NIK(+)EC were present, but this was not predictive of the development of arthritis. CONCLUSION NIK(+)EC are present in the earliest phase of synovial inflammation and may be indicative of high angiogenic activity in the inflamed ST. Therefore, NIK(+)EC may play an important role in the persistence of synovitis. Collectively, our data underscore the importance of angiogenesis in synovial inflammation and identify NIK as a potential therapeutic target in arthritis.
Collapse
Affiliation(s)
- Karen I Maijer
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Ae Ri Noort
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Maria J H de Hair
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Christiaan van der Leij
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Katinka P M van Zoest
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Ivy Y Choi
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Daniëlle M Gerlag
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Mario Maas
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Paul P Tak
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Sander W Tas
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam.
| |
Collapse
|
43
|
Jane-wit D, Surovtseva YV, Qin L, Li G, Liu R, Clark P, Manes TD, Wang C, Kashgarian M, Kirkiles-Smith NC, Tellides G, Pober JS. Complement membrane attack complexes activate noncanonical NF-κB by forming an Akt+ NIK+ signalosome on Rab5+ endosomes. Proc Natl Acad Sci U S A 2015; 112:9686-91. [PMID: 26195760 PMCID: PMC4534258 DOI: 10.1073/pnas.1503535112] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Complement membrane attack complexes (MACs) promote inflammatory functions in endothelial cells (ECs) by stabilizing NF-κB-inducing kinase (NIK) and activating noncanonical NF-κB signaling. Here we report a novel endosome-based signaling complex induced by MACs to stabilize NIK. We found that, in contrast to cytokine-mediated activation, NIK stabilization by MACs did not involve cIAP2 or TRAF3. Informed by a genome-wide siRNA screen, instead this response required internalization of MACs in a clathrin-, AP2-, and dynamin-dependent manner into Rab5(+)endosomes, which recruited activated Akt, stabilized NIK, and led to phosphorylation of IκB kinase (IKK)-α. Active Rab5 was required for recruitment of activated Akt to MAC(+) endosomes, but not for MAC internalization or for Akt activation. Consistent with these in vitro observations, MAC internalization occurred in human coronary ECs in vivo and was similarly required for NIK stabilization and EC activation. We conclude that MACs activate noncanonical NF-κB by forming a novel Akt(+)NIK(+) signalosome on Rab5(+) endosomes.
Collapse
Affiliation(s)
- Dan Jane-wit
- Division of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Yulia V Surovtseva
- Yale Center for Molecular Discovery, Yale University, New Haven, CT 06516
| | - Lingfeng Qin
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06520
| | - Guangxin Li
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06520
| | - Rebecca Liu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Pamela Clark
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Thomas D Manes
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Chen Wang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Michael Kashgarian
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520
| | | | - George Tellides
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06520
| | - Jordan S Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520; Department of Pathology, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
44
|
Wan X, Hou LJ, Zhang LY, Huang WJ, Liu L, Zhang Q, Hu B, Chen W, Chen X, Cao CC. IKKα is involved in kidney recovery and regeneration of acute ischemia/reperfusion injury in mice through IL10-producing regulatory T cells. Dis Model Mech 2015; 8:733-42. [PMID: 26035380 PMCID: PMC4486855 DOI: 10.1242/dmm.018200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 04/17/2015] [Indexed: 12/30/2022] Open
Abstract
The recovery phase after kidney ischemia/reperfusion (IR) injury is often associated with the suppression of inflammation and the proliferation of tubular epithelial cells (TECs). The duration of this phase is often determined by the suppression of inflammation and the proliferation of TECs. Several lines of evidence suggest that IκB kinase α (IKKα) not only promotes the production of anti-inflammatory factors and/or prevents the production of inflammatory factors, but also induces the accompanying cell differentiation and regeneration, and suppresses inflammation. We therefore hypothesized that IKKα could participate in the kidney repair after IR injury and have used a mouse model of acute kidney injury (AKI) to test this. We found that IKKα mediated the repair of the kidney via infiltrated regulatory T (Treg) cells, which can produce anti-inflammatory cytokine IL10, and that IKKα also increased the proliferation of the surviving TECs and suppressed of inflammation. In addition, the expression of indoleamine 2,3-dioxygenase (IDO) in TECs is consistent with the infiltration of IL10-producing Treg cells. We conclude that IKKα is involved in kidney recovery and regeneration through the Treg cells that can produce IL10, which might be a potential therapeutic target that can be used to promote kidney repair after IR injury.
Collapse
Affiliation(s)
- Xin Wan
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Li-Jun Hou
- Division of Neurosurgery, Department of Surgery and Shanghai Neurosurgical Institute, The Second Military Medical University, Changzheng Hospital, Shanghai 200003, China
| | - Li-Yuan Zhang
- Division of Nephrology, Department of Medicine, Affiliated Lianyungang Clinical Medical College of Nanjing Medical University, Lianyungang 222002, China
| | - Wen-Juan Huang
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Lin Liu
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Qian Zhang
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Bo Hu
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Wen Chen
- Division of Cardiovascular Surgery, Department of Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xin Chen
- Division of Cardiovascular Surgery, Department of Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Chang-Chun Cao
- Division of Nephrology, Department of Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
45
|
McCullagh BN, Costello CM, Li L, O’Connell C, Codd M, Lawrie A, Morton A, Kiely DG, Condliffe R, Elliot C, McLoughlin P, Gaine S. Elevated plasma CXCL12α is associated with a poorer prognosis in pulmonary arterial hypertension. PLoS One 2015; 10:e0123709. [PMID: 25856504 PMCID: PMC4391833 DOI: 10.1371/journal.pone.0123709] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/05/2015] [Indexed: 01/24/2023] Open
Abstract
RATIONALE Recent work in preclinical models suggests that signalling via the pro-angiogenic and pro-inflammatory cytokine, CXCL12 (SDF-1), plays an important pathogenic role in pulmonary hypertension (PH). The objective of this study was to establish whether circulating concentrations of CXCL12α were elevated in patients with PAH and related to mortality. METHODS Plasma samples were collected from patients with idiopathic pulmonary arterial hypertension (IPAH) and PAH associated with connective tissue diseases (CTD-PAH) attending two pulmonary hypertension referral centres (n = 95) and from age and gender matched healthy controls (n = 44). Patients were subsequently monitored throughout a period of five years. RESULTS CXCL12α concentrations were elevated in PAH groups compared to controls (P<0.05) and receiver-operating-characteristic analysis showed that plasma CXCL12α concentrations discriminated patients from healthy controls (AUC 0.80, 95% confidence interval 0.73-0.88). Kaplan Meier analysis indicated that elevated plasma CXCL12α concentration was associated with reduced survival (P<0.01). Multivariate Cox proportional hazards model showed that elevated CXCL12α independently predicted (P<0.05) earlier death in PAH with a hazard ratio (95% confidence interval) of 2.25 (1.01-5.00). In the largest subset by WHO functional class (Class 3, 65% of patients) elevated CXCL12α independently predicted (P<0.05) earlier death, hazard ratio 2.27 (1.05-4.89). CONCLUSIONS Our data show that elevated concentrations of circulating CXCL12α in PAH predicted poorer survival. Furthermore, elevated circulating CXCL12α was an independent risk factor for death that could potentially be included in a prognostic model and guide therapy.
Collapse
Affiliation(s)
- Brian N. McCullagh
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
- Pulmonary Hypertension Unit, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Christine M. Costello
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Lili Li
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Caroline O’Connell
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
- Pulmonary Hypertension Unit, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Mary Codd
- School of Public Health, Physiotherapy and Population Science, University College Dublin, Dublin 4, Ireland
| | - Allan Lawrie
- Department of Cardiovascular Sciences, University of Sheffield, Sheffield, United Kingdom
| | - Allison Morton
- Department of Cardiology, Northern General Hospital, Sheffield, United Kingdom
| | - David G. Kiely
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Robin Condliffe
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Charles Elliot
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Paul McLoughlin
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
- * E-mail:
| | - Sean Gaine
- Pulmonary Hypertension Unit, Mater Misericordiae University Hospital, Dublin 7, Ireland
| |
Collapse
|
46
|
Zhang H, Teng X, Liu Z, Zhang L, Liu Z. Gene expression profile analyze the molecular mechanism of CXCR7 regulating papillary thyroid carcinoma growth and metastasis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:16. [PMID: 25887589 PMCID: PMC4349308 DOI: 10.1186/s13046-015-0132-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 02/02/2015] [Indexed: 11/30/2022]
Abstract
Background To detect genetic expression profile alterations after papillary thyroid carcinoma (PTC) cells transfected with chemokine receptor CXCR7 gene by gene microarray, and gain insights into molecular mechanisms of how CXCR7 regulating PTC growth and metastasis. Methods The Human OneArray microarray was used for a complete genome-wide transcript profiling of CXCR7 transfected PTCs (K1-CXCR7 cells), defined as experimental group. Non CXCR7 transfected PTCs (K1 cells) were used as control group. Differential analysis for per gene was performed with a random variance model and t test, p values were adjusted to control the false discovery rate. Gene ontology (GO) on differentially expressed genes to identify the biological processes in modulating the progression of papillary thyroid carcinoma. Pathway analysis was used to evaluate the signaling pathway that differentially expressed genes were involved in. In addition, quantitative real-time polymerase chain reaction (q-PCR) and Western blot were used to verify the top differentially expression genes. Results Comparative analysis revealed that the expression level of 1149 genes was changed in response to CXCR7 transfection. After unsupervised hierarchical clustering analysis, 270 differentially expressed genes were filtered, of them 156 genes were up-regulated whereas 114 genes were down-regulated in K1-CXCR7 cells. GO enrichment analysis revealed the differentially expressed genes were mainly involved in biopolymer metabolic process, signal transduction and protein metabolism. Pathway enrichment analysis revealed differentially expressed genes were mainly involved in ECM-receptor interaction, Focal adhesion, MAPK signaling pathway and Cytokine-cytokine receptor interaction pathway. More importantly, the expression level of genes closely associated with tumor growth and metastasis was altered significantly in K1-CXCR7 cells, including up-regulated genes FN1, COL1A1, COL4A1, PDGFRB, LTB, CXCL12, MMP-11, MT1-MMP and down-regulated genes ITGA7, and Notch-1. Conclusions Gene expression profiling analysis of papillary thyroid carcinoma can further delineate the mechanistic insights on how CXCR7 regulating papillary thyroid carcinoma growth and metastasis. CXCR7 may regulate growth and metastasis of papillary thyroid carcinoma via the activation of PI3K/AKT pathway and its downstream NF-κB signaling, as well as the down-regulation of Notch signaling.
Collapse
Affiliation(s)
- Hengwei Zhang
- Department General Surgery, Affiliated Shenjing Hospital, China Medical University, No.36 Sanhao Street, Shenyang, 110004, China.
| | - Xuyong Teng
- Department General Surgery, Affiliated Shenjing Hospital, China Medical University, No.36 Sanhao Street, Shenyang, 110004, China.
| | - Zhangyi Liu
- Department General Surgery, Affiliated Shenjing Hospital, China Medical University, No.36 Sanhao Street, Shenyang, 110004, China.
| | - Lei Zhang
- Department General Surgery, Affiliated Shenjing Hospital, China Medical University, No.36 Sanhao Street, Shenyang, 110004, China.
| | - Zhen Liu
- Department General Surgery, Affiliated Shenjing Hospital, China Medical University, No.36 Sanhao Street, Shenyang, 110004, China.
| |
Collapse
|
47
|
Noort AR, Tak PP, Tas SW. Non-canonical NF-κB signaling in rheumatoid arthritis: Dr Jekyll and Mr Hyde? Arthritis Res Ther 2015; 17:15. [PMID: 25774937 PMCID: PMC4308835 DOI: 10.1186/s13075-015-0527-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The nuclear factor-κB (NF-κB) family of transcription factors is essential for the expression of pro-inflammatory cytokines, but can also induce regulatory pathways. NF-κB can be activated via two distinct pathways: the classical or canonical pathway, and the alternative or non-canonical pathway. It is well established that the canonical NF-κB pathway is essential both in acute inflammatory responses and in chronic inflammatory diseases, including rheumatoid arthritis (RA). Although less extensively studied, the non-canonical NF-κB pathway is not only central in lymphoid organ development and adaptive immune responses, but is also thought to play an important role in the pathogenesis of RA. Importantly, this pathway appears to have cell type-specific functions and, since many different cell types are involved in the pathogenesis of RA, it is difficult to predict the net overall contribution of the non-canonical NF-κB pathway to synovial inflammation. In this review, we describe the current understanding of non-canonical NF-κB signaling in various important cell types in the context of RA and consider the relevance to the pathogenesis of the disease. In addition, we discuss current drugs targeting this pathway, as well as future therapeutic prospects.
Collapse
|
48
|
Licensed human natural killer cells aid dendritic cell maturation via TNFSF14/LIGHT. Proc Natl Acad Sci U S A 2014; 111:E5688-96. [PMID: 25512551 DOI: 10.1073/pnas.1411072112] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Interactions between natural killer (NK) cells and dendritic cells (DCs) aid DC maturation and promote T-cell responses. Here, we have analyzed the response of human NK cells to tumor cells, and we identify a pathway by which NK-DC interactions occur. Gene expression profiling of tumor-responsive NK cells identified the very rapid induction of TNF superfamily member 14 [TNFSF14; also known as homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for HVEM, a receptor expressed by T lymphocytes (LIGHT)], a cytokine implicated in the enhancement of antitumor responses. TNFSF14 protein expression was induced by three primary mechanisms of NK cell activation, namely, via the engagement of CD16, by the synergistic activity of multiple target cell-sensing NK-cell activation receptors, and by the cytokines IL-2 and IL-15. For antitumor responses, TNFSF14 was preferentially produced by the licensed NK-cell population, defined by the expression of inhibitory receptors specific for self-MHC class I molecules. In contrast, IL-2 and IL-15 treatment induced TNFSF14 production by both licensed and unlicensed NK cells, reflecting the ability of proinflammatory conditions to override the licensing mechanism. Importantly, both tumor- and cytokine-activated NK cells induced DC maturation in a TNFSF14-dependent manner. The coupling of TNFSF14 production to tumor-sensing NK-cell activation receptors links the tumor immune surveillance function of NK cells to DC maturation and adaptive immunity. Furthermore, regulation by NK cell licensing helps to safeguard against TNFSF14 production in response to healthy tissues.
Collapse
|
49
|
Noort AR, van Zoest KPM, Weijers EM, Koolwijk P, Maracle CX, Novack DV, Siemerink MJ, Schlingemann RO, Tak PP, Tas SW. NF-κB-inducing kinase is a key regulator of inflammation-induced and tumour-associated angiogenesis. J Pathol 2014; 234:375-85. [PMID: 25043127 PMCID: PMC4194146 DOI: 10.1002/path.4403] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/23/2014] [Accepted: 07/02/2014] [Indexed: 01/08/2023]
Abstract
Angiogenesis is essential during development and in pathological conditions such as chronic inflammation and cancer progression. Inhibition of angiogenesis by targeting vascular endothelial growth factor (VEGF) blocks disease progression, but most patients eventually develop resistance which may result from compensatory signalling pathways. In endothelial cells (ECs), expression of the pro-angiogenic chemokine CXCL12 is regulated by non-canonical nuclear factor (NF)-κB signalling. Here, we report that NF-κB-inducing kinase (NIK) and subsequent non-canonical NF-κB signalling regulate both inflammation-induced and tumour-associated angiogenesis. NIK is highly expressed in endothelial cells (ECs) in tumour tissues and inflamed rheumatoid arthritis synovial tissue. Furthermore, non-canonical NF-κB signalling in human microvascular ECs significantly enhanced vascular tube formation, which was completely blocked by siRNA targeting NIK. Interestingly, Nik(-/-) mice exhibited normal angiogenesis during development and unaltered TNFα- or VEGF-induced angiogenic responses, whereas angiogenesis induced by non-canonical NF-κB stimuli was significantly reduced. In addition, angiogenesis in experimental arthritis and a murine tumour model was severely impaired in these mice. These studies provide evidence for a role of non-canonical NF-κB signalling in pathological angiogenesis, and identify NIK as a potential therapeutic target in chronic inflammatory diseases and tumour neoangiogenesis.
Collapse
Affiliation(s)
- Ae R Noort
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Katinka PM van Zoest
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Ester M Weijers
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU Medical CenterAmsterdam, The Netherlands
| | - Pieter Koolwijk
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU Medical CenterAmsterdam, The Netherlands
| | - Chrissta X Maracle
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Deborah V Novack
- Division of Bone and Mineral Diseases, Departments of Medicine and Pathology, Washington University School of MedicineSt Louis, Missouri, USA
| | - Martin J Siemerink
- Ocular Angiogenesis Group, Department of Ophthalmology and Department of Cell Biology and Histology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Department of Ophthalmology and Department of Cell Biology and Histology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Paul P Tak
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Medicine, University of CambridgeCambridge, UK
| | - Sander W Tas
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of AmsterdamThe Netherlands
| |
Collapse
|
50
|
Choi JS, Park BC, Chi SW, Bae KH, Kim S, Cho S, Son WC, Myung PK, Kim JH, Park SG. HAX1 regulates E3 ubiquitin ligase activity of cIAPs by promoting their dimerization. Oncotarget 2014; 5:10084-99. [PMID: 25275296 PMCID: PMC4259407 DOI: 10.18632/oncotarget.2459] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 09/06/2014] [Indexed: 12/03/2022] Open
Abstract
HS-1-associated protein X-1 (HAX1) is a multi-functional protein which was first identified as a Hematopoietic cell specific Lyn Substrate 1 (HS1)-binding protein. Although the roles of HAX1 in apoptosis have been unraveled and HAX1 has been proposed to be involved in several diseases, additional roles of HAX1 are still being identified. Here, we demonstrated that HAX1 directly interacted with cellular Inhibitor of Apoptosis Proteins (cIAPs), ubiquitin E3 ligases which regulate the abundance of cellular proteins, via ubiquitin-dependent proteasomal degradation. We showed that HAX1 promotes auto-ubiquitination and degradation of cIAPs by facilitating the intermolecular homodimerization of RING finger domain. Moreover, HAX1 regulates the non-canonical Nuclear Factor-κB (NF-κB) signaling pathway by modulating the stability of NF-κB-Inducing Kinase (NIK), which is one of the substrates of cIAPs. Taken together, these results unveil a novel role of HAX1 in the non-canonical NF-κB pathway, and provide an important clue that HAX1 is a potential therapeutic target for the treatment of cancer.
Collapse
Affiliation(s)
- Jin Sun Choi
- Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Byoung Chul Park
- Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Seung Wook Chi
- Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Kwang-Hee Bae
- Cell Function Regulation Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Sunhong Kim
- Targeted Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Biomolecular Science, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Sayeon Cho
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Woo-Chan Son
- Asan Institute for Life Sciences and Asan Medical Center, Seoul, Republic of Korea
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Pyung Keun Myung
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Jeong-Hoon Kim
- Targeted Gene Regulation Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, Daejeon, Republic of Korea
| | - Sung Goo Park
- Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
| |
Collapse
|