1
|
Lee KY, Mei Y, Liu H, Schwarz H. CD137-expressing regulatory T cells in cancer and autoimmune diseases. Mol Ther 2025; 33:51-70. [PMID: 39668561 PMCID: PMC11764688 DOI: 10.1016/j.ymthe.2024.12.010] [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: 09/07/2024] [Revised: 11/25/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024] Open
Abstract
Regulatory T cells (Tregs) are essential for maintaining immune homeostasis, with critical roles in preventing aberrant immune responses that occur in autoimmune diseases and chronic inflammation. Conversely, the abundance of Tregs in cancer is associated with impaired anti-tumor immunity, and tumor immune evasion. Recent work demonstrates that CD137, a well-known costimulatory molecule for T cells, is highly expressed on Tregs in pathological conditions, while its expression is minimal or negligible on peripheral Tregs. The expression of CD137 marks Tregs with potent immunosuppressive phenotype that foster cancer progression and are protective against certain autoimmune diseases. Hence CD137 has emerged as a marker for Tregs. However, several important questions still remain regarding the expression and function of CD137 in Tregs. Here, we provide an overview of our current knowledge of Treg mechanisms of action, with a focus on the role of CD137 in modulating Treg activity. We also explore the implications of CD137+ Tregs in both cancer and autoimmune diseases, emphasizing the significance of targeting these cells for therapeutic intervention in these conditions.
Collapse
Affiliation(s)
- Kang Yi Lee
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore
| | - Yu Mei
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore
| | - Haiyan Liu
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore.
| | - Herbert Schwarz
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore.
| |
Collapse
|
2
|
Yi L, Yan J, Wei P, Long S, Wang X, Gu M, Yang B, Chen Y, Ma S, Wang C, Zheng M, Sun Q, Shi Y, Wang G. The levels of soluble CD137 are increased in tuberculosis patients and associated with disease severity and prognosis. Eur J Immunol 2024; 54:e2350796. [PMID: 38922884 DOI: 10.1002/eji.202350796] [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: 09/29/2023] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 06/28/2024]
Abstract
Tuberculosis (TB) was the leading cause of death from a single infectious agent before the coronavirus pandemic. Therefore, it is important to search for severity biomarkers and devise appropriate therapies. A total of 139 pulmonary TB (PTB) patients and 80 healthy controls (HCs) were recruited for plasma soluble CD137 (sCD137) detection through ELISA. Moreover, pleural effusion sCD137 levels were measured in 85 TB patients and 36 untreated lung cancer patients. The plasma cytokine levels in 64 patients with PTB and blood immune cell subpopulations in 68 patients with PTB were analysed via flow cytometry. Blood sCD137 levels were higher in PTB patients (p = 0.012) and correlated with disease severity (p = 0.0056). The level of sCD137 in tuberculous pleurisy effusion (TPE) was markedly higher than that in malignant pleurisy effusion (p = 0.018). Several blood cytokines, such as IL-6 (p = 0.0147), IL-8 (p = 0.0477), IP-10 (p ≤ 0.0001) and MCP-1 (p = 0.0057), and some laboratory indices were significantly elevated in severe PTB (SE) patients, but the percentages of total lymphocytes (p = 0.002) and cytotoxic T cells (p = 0.036) were significantly lower in SE patients than in non-SE patients. In addition, the sCD137 level was negatively correlated with the percentage of total lymphocytes (p = 0.0008) and cytotoxic T cells (p = 0.0021), and PTB patients with higher plasma sCD137 levels had significantly shorter survival times (p = 0.0041). An increase in sCD137 is a potential biomarker for severe TB and indicates a poor prognosis.
Collapse
Affiliation(s)
- Ling Yi
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jun Yan
- Department of Clinical Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Panjian Wei
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Sibo Long
- Department of Clinical Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xiaojue Wang
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Meng Gu
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Bin Yang
- Department of Clinical Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Yan Chen
- Department of Clinical Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Shang Ma
- Department of Clinical Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Chaohong Wang
- Department of Clinical Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Maike Zheng
- Department of Clinical Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Qing Sun
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Yiheng Shi
- Department of Clinical Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Guirong Wang
- Department of Clinical Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Houlder EL, Stam KA, Koopman JPR, König MH, Langenberg MCC, Hoogerwerf MA, Niewold P, Sonnet F, Janse JJ, Partal MC, Sijtsma JC, de Bes-Roeleveld LHM, Kruize YCM, Yazdanbakhsh M, Roestenberg M. Early symptom-associated inflammatory responses shift to type 2 responses in controlled human schistosome infection. Sci Immunol 2024; 9:eadl1965. [PMID: 38968336 DOI: 10.1126/sciimmunol.adl1965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 06/07/2024] [Indexed: 07/07/2024]
Abstract
Schistosomiasis is an infection caused by contact with Schistosoma-contaminated water and affects more than 230 million people worldwide with varying morbidity. The roles of T helper 2 (TH2) cells and regulatory immune responses in chronic infection are well documented, but less is known about human immune responses during acute infection. Here, we comprehensively map immune responses during controlled human Schistosoma mansoni infection using male or female cercariae. Immune responses to male or female parasite single-sex infection were comparable. An early TH1-biased inflammatory response was observed at week 4 after infection, which was particularly apparent in individuals experiencing symptoms of acute schistosomiasis. By week 8 after infection, inflammatory responses were followed by an expansion of TH2 and regulatory cell subsets. This study demonstrates the shift from TH1 to both TH2 and regulatory responses, typical of chronic schistosomiasis, in the absence of egg production and provides immunological insight into the clinical manifestations of acute schistosomiasis.
Collapse
Affiliation(s)
- Emma L Houlder
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Koen A Stam
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Jan Pieter R Koopman
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Marion H König
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Marijke C C Langenberg
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Marie-Astrid Hoogerwerf
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Paula Niewold
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Friederike Sonnet
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Jacqueline J Janse
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Miriam Casacuberta Partal
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Jeroen C Sijtsma
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Laura H M de Bes-Roeleveld
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Yvonne C M Kruize
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Maria Yazdanbakhsh
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Meta Roestenberg
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| |
Collapse
|
4
|
Yuan L, Wang Y, Shen X, Ma F, Wang J, Yan F. Soluble form of immune checkpoints in autoimmune diseases. J Autoimmun 2024; 147:103278. [PMID: 38943864 DOI: 10.1016/j.jaut.2024.103278] [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: 02/01/2024] [Revised: 05/03/2024] [Accepted: 06/19/2024] [Indexed: 07/01/2024]
Abstract
Immune checkpoints are essential regulators of immune responses, either by activating or suppressing them. Consequently, they are regarded as pivotal elements in the management of infections, cancer, and autoimmune disorders. In recent years, researchers have identified numerous soluble immune checkpoints that are produced through various mechanisms and demonstrated biological activity. These soluble immune checkpoints can be produced and distributed in the bloodstream and various tissues, with their roles in immune response dysregulation and autoimmunity extensively documented. This review aims to provide a thorough overview of the generation of various soluble immune checkpoints, such as sPD-1, sCTLA-4, sTim-3, s4-1BB, sBTLA, sLAG-3, sCD200, and the B7 family, and their importance as indicators for the diagnosis and prediction of autoimmune conditions. Furthermore, the review will investigate the potential pathological mechanisms of soluble immune checkpoints in autoimmune diseases, emphasizing their association with autoimmune diseases development, prognosis, and treatment.
Collapse
Affiliation(s)
- Li Yuan
- Geriatric Diseases Institute of Chengdu, Department of Clinical Laboratory, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China
| | - Yuxia Wang
- Geriatric Intensive Care Unit, Sichuan Geriatric Medical Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Xuxia Shen
- Geriatric Diseases Institute of Chengdu, Department of Clinical Laboratory, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China
| | - Fujun Ma
- Department of Training, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China
| | - Jun Wang
- Department of Respiratory and Critical Care Medicine, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China.
| | - Fang Yan
- Geriatric Diseases Institute of Chengdu, Department of Geriatrics, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China; Geriatric Diseases Institute of Chengdu, Department of Intensive Care Medicine, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China; Center for Medicine Research and Translation, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China.
| |
Collapse
|
5
|
Reiter A, Verweyen EL, Queste E, Fuehner S, Jakob A, Masjosthusmann K, Hinze C, Wittkowski H, Foell D, Meinzer U, Melki I, Kessel C. Proteomic mapping identifies serum marker signatures associated with MIS-C specific hyperinflammation and cardiovascular manifestation. Clin Immunol 2024; 264:110237. [PMID: 38723855 DOI: 10.1016/j.clim.2024.110237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/19/2024]
Abstract
Multisystem inflammatory syndrome in children (MIS-C) shares several clinical and immunological features with Kawasaki Disease (KD) and pediatric hyperinflammation, but the immuno-phenotypic overlap among these clinical mimics is still incompletely understood. Here we analyzed serum samples from treatment-naïve patients with MIS-C (n = 31) and KD (n = 11), pediatric hyperinflammation (n = 13) and healthy controls (HC, n = 10) by proximity extension assay (PEA) to profile 184 blood biomarkers. Collectively, immunophenotypic overlap between MIS-C and hyperinflammation exceeds overlap with KD. Overexpression of IL-17A in MIS-C and KD could best separate these conditions from hyperinflammatory conditions, while those were hallmarked by overabundance of adenosin deaminase and IL-18. Depletion in serum TNF-related subfamily member 9 (TNFRSF9) and apoptosis inducing ligand (TRAIL) linked with cardiovascular manifestations and myocarditis in MIS-C. Altogether, our analysis highlights important differences in molecular marker signatures also across different MIS-C and KD cohorts and suggests several previously unidentified molecular associations in context of cardiovascular inflammation.
Collapse
Affiliation(s)
- Andrea Reiter
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany
| | - Emely L Verweyen
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany
| | - Emmanuelle Queste
- Department of General Pediatrics, Pediatric Internal Medicine, Rheumatology and Infectious Diseases, National Reference Centre for Rare Pediatric Inflammatory Rheumatisms and Systemic Autoimmune diseases (RAISE), Robert-Debré University Hospital, Assistance Publique-Hôpitaux de Paris, F-75019 Paris, France; Université Paris Cité, INSERM, Centre de Recherche sur l'inflammation UMR 1149, Paris, France
| | - Sabrina Fuehner
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany
| | - André Jakob
- Division of Pediatric Cardiology and Pediatric Intensive Care, Ludwig-Maximilians University, Munich, Germany
| | - Katja Masjosthusmann
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster, Germany
| | - Claas Hinze
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany
| | - Helmut Wittkowski
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany
| | - Dirk Foell
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany
| | - Ulrich Meinzer
- Department of General Pediatrics, Pediatric Internal Medicine, Rheumatology and Infectious Diseases, National Reference Centre for Rare Pediatric Inflammatory Rheumatisms and Systemic Autoimmune diseases (RAISE), Robert-Debré University Hospital, Assistance Publique-Hôpitaux de Paris, F-75019 Paris, France; Université Paris Cité, INSERM, Centre de Recherche sur l'inflammation UMR 1149, Paris, France
| | - Isabelle Melki
- Department of General Pediatrics, Pediatric Internal Medicine, Rheumatology and Infectious Diseases, National Reference Centre for Rare Pediatric Inflammatory Rheumatisms and Systemic Autoimmune diseases (RAISE), Robert-Debré University Hospital, Assistance Publique-Hôpitaux de Paris, F-75019 Paris, France; Paediatrics, Rheumatology and Paediatric Internal Medicine, Children's Hospital, F-33000 Bordeaux, France; Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Université Paris Cité, Inserm UMR 1163, F-75015 Paris, France
| | - Christoph Kessel
- Department of Pediatric Rheumatology & Immunology, University Children's Hospital, Muenster, Germany.
| |
Collapse
|
6
|
Lima CAC, Martins MR, Dos Santos RL, da Silva LM, Da Silva JPA, Forones NM, Torres LC. Soluble levels of 4-1BB (CD137) and OX40 (CD134) are associated with cancer progression in gastric adenocarcinoma. J Surg Oncol 2024. [PMID: 38853545 DOI: 10.1002/jso.27726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND AND OBJECTIVES Previous studies have demonstrated that soluble forms of T-cell costimulatory molecules 4-1BB (s4-1BB) and OX40 (sOX40) interact with immune cells and may constitute a mechanism of immune evasion by tumors in various cancers. The role of the soluble forms of 4-1BB and OX40 in GC remains unclear. We aimed to examine the association between serum levels of s4-1BB and sOX40 and tumor progression in patients with GC. METHODS Between 2017 and 2018, a cross-sectional study was performed with serum samples of 83 GC patients and 20 healthy controls. RESULTS Patients with stage IV metastatic gastric cancer had significantly higher levels of soluble OX40 in comparison with stage III patients with lymph nodes metastasis (p = 0.0003) and stages I and II patients (p = 0.005), whereas the opposite was found for soluble 4-1BB levels, with lower levels being found in advanced stage III (p = 0.003) compared with initial stages I/II. CONCLUSIONS The sOX40 and s4-1BB-mediated T cell interactions may be involved in antitumor immune responses in GC, possibly favoring tumor escape and progression. Serum levels of sOX40 and s4-1BB are associated with staging in GC and may constitute biomarkers for prognosis, as well as potential targets for immunotherapy.
Collapse
Affiliation(s)
- Cecilia Araújo Carneiro Lima
- Translational Research Laboratory Prof CA Hart (IMIP), Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Hospital de Câncer de Pernambuco, Recife, Brazil
- Postgraduate program in translational medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- Real Instituto de Cirurgia Oncológica (RICO-RHP), Recife, Brazil
| | - Mário Rino Martins
- Translational Research Laboratory Prof CA Hart (IMIP), Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Hospital de Câncer de Pernambuco, Recife, Brazil
- Real Instituto de Cirurgia Oncológica (RICO-RHP), Recife, Brazil
| | - Rogerio Luiz Dos Santos
- Translational Research Laboratory Prof CA Hart (IMIP), Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Hospital de Câncer de Pernambuco, Recife, Brazil
- Real Instituto de Cirurgia Oncológica (RICO-RHP), Recife, Brazil
| | - Luciana Mata da Silva
- Translational Research Laboratory Prof CA Hart (IMIP), Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Hospital de Câncer de Pernambuco, Recife, Brazil
- Postgraduate program in translational medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Jeronimo Paulo Assis Da Silva
- Translational Research Laboratory Prof CA Hart (IMIP), Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Hospital de Câncer de Pernambuco, Recife, Brazil
- Postgraduate program in translational medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- Real Instituto de Cirurgia Oncológica (RICO-RHP), Recife, Brazil
| | - Nora Manoukian Forones
- Department of Digestive Surgery, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Leuridan Cavalcante Torres
- Translational Research Laboratory Prof CA Hart (IMIP), Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Hospital de Câncer de Pernambuco, Recife, Brazil
- Postgraduate program in translational medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
7
|
Maddaloni E, Amendolara R, Balena A, Latino A, Sessa RL, Buzzetti R. Immune checkpoint modulators in early clinical development for the treatment of type 1 diabetes. Expert Opin Investig Drugs 2024; 33:303-318. [PMID: 38427915 DOI: 10.1080/13543784.2024.2326036] [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: 09/28/2023] [Accepted: 02/28/2024] [Indexed: 03/03/2024]
Abstract
INTRODUCTION Despite the improvements of insulin therapy, people with type 1 diabetes (T1D) still suffer from a decreased quality of life and life expectancy. The search toward a cure for T1D is therefore still a scorching open field of research. AREAS COVERED Tackling the immune checkpoint signaling pathways has gained importance in the field of cancer immunotherapy. The same pathways can be targeted in autoimmunity with an opposite principle: to dampen the exaggerated immune response. In this review, we report a comprehensive excursus on the cellular and molecular mechanisms that lead to loss of immunological tolerance, and recent evidence on the role of immune checkpoint molecules in the development of T1D and their potential application for the mitigation of autoimmune diabetes. EXPERT OPINION Contrasting results about the efficacy of immune checkpoint modulators for T1D have been published, with very few molecules from preclinical studies eligible for use in humans. The heterogeneous and complex pathophysiology of T1D may explain the conflicting evidence. Designing clinical trials that acknowledge the pathophysiological and clinical complexity of T1D and that forecast the need of simultaneously tackling different disease pathways will be crucial to enhance the benefits which may be gained by such compounds.
Collapse
Affiliation(s)
- Ernesto Maddaloni
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Rocco Amendolara
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Angela Balena
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandro Latino
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Rosario Luigi Sessa
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Raffaella Buzzetti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
8
|
Bruzzaniti S, Piemonte E, Bruzzese D, Lepore MT, Strollo R, Izzo L, Di Candia F, Franzese A, Bifulco M, Mozzillo E, Ludvigsson J, Matarese G, Galgani M. Progression of type 1 diabetes is associated with high levels of soluble PD-1 in islet autoantibody-positive children. Diabetologia 2024; 67:714-723. [PMID: 38214712 PMCID: PMC10904438 DOI: 10.1007/s00125-023-06075-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/27/2023] [Indexed: 01/13/2024]
Abstract
AIMS/HYPOTHESIS Type 1 diabetes is an autoimmune disorder that is characterised by destruction of pancreatic beta cells by autoreactive T lymphocytes. Although islet autoantibodies (AAb) are an indicator of disease progression, specific immune biomarkers that can be used as target molecules to halt development of type 1 diabetes have not been discovered. Soluble immune checkpoint molecules (sICM) play a pivotal role in counteracting excessive lymphocyte responses, but their role in type 1 diabetes is unexplored. In this longitudinal study, we measured sICM levels in AAb-positive (AAb+) children to identify molecules related to type 1 diabetes progression. METHODS We measured the levels of 14 sICM in the sera of AAb+ children (n=57) compared to those with recent-onset type 1 diabetes (n=79) and healthy children (n=44), obtained from two cohorts. AAb+ children were followed up and divided based on their progression to type 1 diabetes (AAbP) or not (AAbNP) (if they lost islet autoimmunity and did not develop disease in subsequent years). sICM were also measured in the sample taken at the visit closest to disease onset in AAbP children. RESULTS We found that AAb+ children had a distinct sICM profile compared with healthy children and those with recent-onset type 1 diabetes. In addition, AAb+ children who progressed to type 1 diabetes (AAbP) had higher sICM concentrations than non-progressors (AAbNP). Further, sICM levels decreased in AAbP children close to disease onset. Application of Cox regression models highlighted that high concentrations of soluble programmed cell death protein 1 (sPD-1) are associated with type 1 diabetes progression (HR 1.71; 95% CI 1.16, 2.51; p=0.007). CONCLUSIONS/INTERPRETATION This study reveals an sICM profile that is dysregulated during the preclinical stage of type 1 diabetes, and identifies sPD-1 as a pathophysiologically-relevant molecule that is associated with disease progression, offering a potential target for early interventions in autoimmune diabetes.
Collapse
Affiliation(s)
- Sara Bruzzaniti
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Erica Piemonte
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Dario Bruzzese
- Dipartimento di Sanità Pubblica, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Maria Teresa Lepore
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Rocky Strollo
- Dipartimento di Scienze Umane e Promozione della Qualità della Vita, Università Telematica San Raffaele Roma, Rome, Italy
| | - Lavinia Izzo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Francesca Di Candia
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Adriana Franzese
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Maurizio Bifulco
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Enza Mozzillo
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Johnny Ludvigsson
- Crown Princess Victoria's Children's Hospital and Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| | - Giuseppe Matarese
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Mario Galgani
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy.
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy.
| |
Collapse
|
9
|
Jin X, Yi L, Wang X, Yan Z, Wei P, Yang B, Zhang H. Costimulatory capacity of CD137 mAbs on T cells depends on elaborate CRD structures but not on blocking ligand-receptor binding. Eur J Immunol 2023; 53:e2350493. [PMID: 37675596 DOI: 10.1002/eji.202350493] [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: 03/18/2023] [Revised: 07/17/2023] [Accepted: 09/05/2023] [Indexed: 09/08/2023]
Abstract
CD137 is mainly a costimulatory receptor of CD8+ T cells. Two representative CD137 antibodies, utomilumab, and urelumab, show different costimulatory capacities in clinical trials. Balancing the antitumor effect and systemic toxicity of T cells activated by CD137 signaling is a challenge that requires clinical consideration. In this study, a panel of specific anti-human CD137 monoclonal antibodies (mAbs) were prepared and their affinities, isotypes, CD137-CRD (cysteine-rich domain) binding regions, cross-reactivity to mouse and rhesus CD137, inhibition of ligand-receptor binding and costimulatory activities were analyzed. The results showed that anti-human CD137 mAbs had high cross-reactivity with rhesus CD137. MAbs fell into three clusters according to their different binding regions of the CD137 extracellular domain. They bound to CRDI+CRDII, CRDIII or CRDIV+STP. CRDIII-binding mAbs had the strongest blocking activity. Highly costimulatory CD137 mAbs showed stronger abilities to promote CD8+ T-cell proliferation. However, the costimulatory capacity of mAbs on T cells was not closely related to their ability to block CD137L-CD137 binding and may be controlled by more elaborate CRD conformational structures. This study provides additional information for the development of next-generation CD137 mAbs to meet clinical needs.
Collapse
Affiliation(s)
- Xin Jin
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumour Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Ling Yi
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumour Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xiaojue Wang
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumour Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Zhuohong Yan
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumour Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Panjian Wei
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumour Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Bin Yang
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumour Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Hongtao Zhang
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumour Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Almenara-Fuentes L, Rodriguez-Fernandez S, Rosell-Mases E, Kachler K, You A, Salvado M, Andreev D, Steffen U, Bang H, Bozec A, Schett G, Le Panse R, Verdaguer J, Dalmases M, Rodriguez-Vidal S, Barneda-Zahonero B, Vives-Pi M. A new platform for autoimmune diseases. Inducing tolerance with liposomes encapsulating autoantigens. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 48:102635. [PMID: 36481472 DOI: 10.1016/j.nano.2022.102635] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/20/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Abstract
Autoimmune diseases (AIDs) are caused by the loss of self-tolerance and destruction of tissues by the host's immune system. Several antigen-specific immunotherapies, focused on arresting the autoimmune attack, have been tested in clinical trials with discouraging results. Therefore, there is a need for innovative strategies to restore self-tolerance safely and definitively in AIDs. We previously demonstrated the therapeutic efficacy of phosphatidylserine (PS)-liposomes encapsulating autoantigens in experimental type 1 diabetes and multiple sclerosis. Here, we show that PS-liposomes can be adapted to other autoimmune diseases by simply replacing the encapsulated autoantigen. After administration, they are distributed to target organs, captured by phagocytes and interact with several immune cells, thus exerting a tolerogenic and immunoregulatory effect. Specific PS-liposomes demonstrate great preventive and therapeutic efficacy in rheumatoid arthritis and myasthenia gravis. Thus, this work highlights the therapeutic potential of a platform for several autoimmunity settings, which is specific, safe, and with long-term effects.
Collapse
Affiliation(s)
| | - Silvia Rodriguez-Fernandez
- Ahead Therapeutics SL, Barcelona, Spain; Immunology Section, Germans Trias i Pujol Research Institute, Autonomous University of Barcelona, Badalona, Spain
| | - Estela Rosell-Mases
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, IRBLleida, University of Lleida, Lleida, Spain
| | - Katerina Kachler
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Axel You
- Sorbonne University, INSERM, Institute of Myology, Center of Research in Myology, F-75013 Paris, France
| | | | - Darja Andreev
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Ulrike Steffen
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | | | - Aline Bozec
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Rozen Le Panse
- Sorbonne University, INSERM, Institute of Myology, Center of Research in Myology, F-75013 Paris, France
| | - Joan Verdaguer
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, IRBLleida, University of Lleida, Lleida, Spain; CIBER of Diabetes and Associated Metabolic Disease (CIBERDEM), ISCIII, Madrid, Spain
| | | | | | | | - Marta Vives-Pi
- Ahead Therapeutics SL, Barcelona, Spain; Immunology Section, Germans Trias i Pujol Research Institute, Autonomous University of Barcelona, Badalona, Spain.
| |
Collapse
|
11
|
Pilśniak A, Otto-Buczkowska E. Type 1 diabetes - What's new in prevention and therapeutic strategies? Pediatr Endocrinol Diabetes Metab 2023; 29:196-201. [PMID: 38031834 PMCID: PMC10679919 DOI: 10.5114/pedm.2023.132028] [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: 06/29/2022] [Accepted: 04/10/2023] [Indexed: 12/01/2023]
Abstract
Type 1 diabetes (T1D) is an autoimmune disorder, and insulin deficiency is the result of b-cell dysfunction. Treatment of type 1 diabetes requires constant parenteral insulin administration, which can be very burdensome for the patient. Meticulous use of insulin therapy does not protect the patient against complications. Hence, the search for other methods of treatment as well as ways of preventing the onset of diabetes has been ongoing for a long time. The main obstacle in the implementation of the prevention task is the need to identify people at risk of developing diabetes before the start of autoimmunity. It seems that primary prevention is still unrealistic at the moment, because we do not know all the factors leading to the activation of autoimmunity processes. Research on the use of late secondary prevention in people who develop glucose tolerance disorders or in the early period after the onset of type 1 diabetes are at the most advanced stage. Gene therapy is another attempt at an alternative treatment and prevention of type 1 diabetes and still requires further research. Recent years have brought a lot of information about the nature of type 1 diabetes and the mechanisms leading to its development. However, it has not yet been established what factors decide about the initiation of autoimmunity and what determines the dynamics of these processes.
Collapse
Affiliation(s)
- Aleksandra Pilśniak
- Department of Internal Medicine, Autoimmune and Metabolic Diseases, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | | |
Collapse
|
12
|
Bruzzaniti S, Piemonte E, Mozzillo E, Bruzzese D, Lepore MT, Carbone F, de Candia P, Strollo R, Porcellini A, Marigliano M, Maffeis C, Bifulco M, Ludvigsson J, Franzese A, Matarese G, Galgani M. High levels of blood circulating immune checkpoint molecules in children with new-onset type 1 diabetes are associated with the risk of developing an additional autoimmune disease. Diabetologia 2022; 65:1390-1397. [PMID: 35610521 DOI: 10.1007/s00125-022-05724-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/01/2022] [Indexed: 11/03/2022]
Abstract
AIMS/HYPOTHESIS We assessed the levels of blood circulating immune checkpoint molecules (ICMs) at diagnosis of type 1 diabetes, and determined their association with the risk of developing an additional autoimmune disorder over time. METHODS Children with new-onset type 1 diabetes (n = 143), without biological and/or clinical signs of additional autoimmune disorders, and healthy children (n = 75) were enrolled, and blood circulating levels of 14 ICMs were measured. The children with type 1 diabetes were divided into two groups on the basis of the development of an additional autoimmune disease in the 5 years after diabetes onset. Differences in soluble ICM levels between the groups were assessed, and a Cox regression analysis was used to evaluate their association with the risk of development of an additional autoimmune disease over time. To validate the data, circulating ICMs were measured in an independent cohort of 60 children with new-onset type 1 diabetes stratified into two groups. RESULTS We found that the levels of circulating ICMs were significantly higher in children with new-onset diabetes compared with healthy children. Further, we observed that children with type 1 diabetes who developed a second autoimmune disease over time (T1D-AAD+ children) had higher levels of soluble ICMs than children with type 1 diabetes who did not (T1D-AAD- children). Cox regression models revealed that high circulating levels of CD137/4-1BB and PD-1 molecules at diabetes diagnosis were associated with the risk of developing an additional autoimmune disease in both type 1 diabetes cohorts. CONCLUSIONS/INTERPRETATION Our findings suggest that soluble CD137/4-1BB and PD-1 molecules may be used as prognostic biomarkers in children with type 1 diabetes, and may pave the way for novel immunological screening at diabetes onset, allowing early identification of children at higher risk of developing other autoimmune conditions over time.
Collapse
Affiliation(s)
- Sara Bruzzaniti
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
- Dipartimento di Biologia, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Erica Piemonte
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Enza Mozzillo
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Dario Bruzzese
- Dipartimento di Sanità Pubblica, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Maria Teresa Lepore
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Fortunata Carbone
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
- Unità di Neuroimmunologia, Fondazione Santa Lucia, Rome, Italy
| | - Paola de Candia
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Rocky Strollo
- Dipartimento di Scienze e Tecnologie per l'Uomo e l'Ambiente, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Antonio Porcellini
- Dipartimento di Biologia, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Marco Marigliano
- Section of Pediatric Diabetes and Metabolism, Department of Surgery, Dentistry, Pediatrics and Gynecology, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Claudio Maffeis
- Section of Pediatric Diabetes and Metabolism, Department of Surgery, Dentistry, Pediatrics and Gynecology, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Maurizio Bifulco
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Johnny Ludvigsson
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, and Crown Princess Victoria Children's Hospital, Region Östergötland, Linköping, Sweden
| | - Adriana Franzese
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Giuseppe Matarese
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Mario Galgani
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy.
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli 'Federico II', Naples, Italy.
| |
Collapse
|
13
|
Rojas M, Heuer LS, Zhang W, Chen YG, Ridgway WM. The long and winding road: From mouse linkage studies to a novel human therapeutic pathway in type 1 diabetes. Front Immunol 2022; 13:918837. [PMID: 35935980 PMCID: PMC9353112 DOI: 10.3389/fimmu.2022.918837] [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: 04/12/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Autoimmunity involves a loss of immune tolerance to self-proteins due to a combination of genetic susceptibility and environmental provocation, which generates autoreactive T and B cells. Genetic susceptibility affects lymphocyte autoreactivity at the level of central tolerance (e.g., defective, or incomplete MHC-mediated negative selection of self-reactive T cells) and peripheral tolerance (e.g., failure of mechanisms to control circulating self-reactive T cells). T regulatory cell (Treg) mediated suppression is essential for controlling peripheral autoreactive T cells. Understanding the genetic control of Treg development and function and Treg interaction with T effector and other immune cells is thus a key goal of autoimmunity research. Herein, we will review immunogenetic control of tolerance in one of the classic models of autoimmunity, the non-obese diabetic (NOD) mouse model of autoimmune Type 1 diabetes (T1D). We review the long (and still evolving) elucidation of how one susceptibility gene, Cd137, (identified originally via linkage studies) affects both the immune response and its regulation in a highly complex fashion. The CD137 (present in both membrane and soluble forms) and the CD137 ligand (CD137L) both signal into a variety of immune cells (bi-directional signaling). The overall outcome of these multitudinous effects (either tolerance or autoimmunity) depends upon the balance between the regulatory signals (predominantly mediated by soluble CD137 via the CD137L pathway) and the effector signals (mediated by both membrane-bound CD137 and CD137L). This immune balance/homeostasis can be decisively affected by genetic (susceptibility vs. resistant alleles) and environmental factors (stimulation of soluble CD137 production). The discovery of the homeostatic immune effect of soluble CD137 on the CD137-CD137L system makes it a promising candidate for immunotherapy to restore tolerance in autoimmune diseases.
Collapse
Affiliation(s)
- Manuel Rojas
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
- School of Medicine and Health Sciences, Doctoral Program in Biological and Biomedical Sciences, Universidad del Rosario, Bogota, Colombia
| | - Luke S. Heuer
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Weici Zhang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Yi-Guang Chen
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Wisconsin, Milwaukee, WI, United States
- Division of Endocrinology, Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, WI, United States
| | - William M. Ridgway
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
- *Correspondence: William M. Ridgway,
| |
Collapse
|
14
|
Serum proteomics links suppression of tumor immunity to ancestry and lethal prostate cancer. Nat Commun 2022; 13:1759. [PMID: 35365620 PMCID: PMC8975871 DOI: 10.1038/s41467-022-29235-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 03/07/2022] [Indexed: 12/31/2022] Open
Abstract
There is evidence that tumor immunobiology and immunotherapy response may differ between African American and European American prostate cancer patients. Here, we determine if men of African descent harbor a unique systemic immune-oncological signature and measure 82 circulating proteins in almost 3000 Ghanaian, African American, and European American men. Protein signatures for suppression of tumor immunity and chemotaxis are elevated in men of West African ancestry. Importantly, the suppression of tumor immunity protein signature associates with metastatic and lethal prostate cancer, pointing to clinical importance. Moreover, two markers, pleiotrophin and TNFRSF9, predict poor disease survival specifically among African American men. These findings indicate that immune-oncology marker profiles differ between men of African and European descent. These differences may contribute to the disproportionate burden of lethal prostate cancer in men of African ancestry. The elevated peripheral suppression of tumor immunity may have important implication for guidance of cancer therapy which could particularly benefit African American patients. Ancestry-related differences in immunobiology may explain the health disparities observed in prostate cancer patients, with men of African origin bearing the highest prostate cancer burden. By measuring immune-related proteins in serum samples, here the authors report that systemic cytokines linked to suppression of tumor immunity are upregulated in men of African ancestry and associated with reduced survival.
Collapse
|
15
|
Yi L, Jin X, Wang J, Yan Z, Cheng X, Wen T, Yang B, Wang X, Che N, Liu Z, Zhang H. CD137 Agonists Targeting CD137-Mediated Negative Regulation Show Enhanced Antitumor Efficacy in Lung Cancer. Front Immunol 2022; 13:771809. [PMID: 35197968 PMCID: PMC8859117 DOI: 10.3389/fimmu.2022.771809] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/19/2022] [Indexed: 11/18/2022] Open
Abstract
Negative immune regulation plays a notable role in tumor immunity. This study aimed to confirm that CD137 mediates negative immunoregulation as well as agonist activity in tumor immunity. Soluble CD137 (sCD137), a prominent splice variant of membrane-bound CD137 (mCD137), was identified, and its concentration in the blood of lung cancer patients was increased. The baseline concentration of sCD137 in the blood was negatively correlated with the efficacy of neoadjuvant immunochemotherapy in a pilot study. The percentage of CD137+ regulatory T cells (Tregs) in the blood of lung cancer patients was also increased, and further enriched at the tumor site; Foxp3, CTLA-4, IL-10, IL-35-Ebi3, sCD137 and costimulatory molecules expression were also higher, indicating increased immunosuppressive activity. A high percentage of CD137+ Tregs in the tumor was associated with worse OS outcomes among patients with high CD137+CD8+ T cell infiltration levels. Notably, targeting CD137+ Tregs using an engineered CD137 agonist with wild-type mouse IgG2a Fc clearly decreased the total Treg numbers and eliminated the tumor in the CT26 model and prolonged the survival rate of a Lewis lung carcinoma (LLC) model. These results indicated it may be possible to empower CD137 agonist with ability to abolish CD137-mediated negative regulation to enhance its antitumor efficacy.
Collapse
Affiliation(s)
- Ling Yi
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xin Jin
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jinghui Wang
- Department of Medical Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Zhuohong Yan
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xu Cheng
- No. 2 Department of Thoracic Surgery, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Tao Wen
- No. 2 Department of Thoracic Surgery, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Bin Yang
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xiaojue Wang
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Nanying Che
- Department of Pathology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Zhidong Liu
- No. 2 Department of Thoracic Surgery, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- *Correspondence: Hongtao Zhang, ; Zhidong Liu,
| | - Hongtao Zhang
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- *Correspondence: Hongtao Zhang, ; Zhidong Liu,
| |
Collapse
|
16
|
Ding JT, Yang KP, Lin KL, Cao YK, Zou F. Mechanisms and therapeutic strategies of immune checkpoint molecules and regulators in type 1 diabetes. Front Endocrinol (Lausanne) 2022; 13:1090842. [PMID: 36704045 PMCID: PMC9871554 DOI: 10.3389/fendo.2022.1090842] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Considered a significant risk to health and survival, type 1 diabetes (T1D) is a heterogeneous autoimmune disease characterized by hyperglycemia caused by an absolute deficiency of insulin, which is mainly due to the immune-mediated destruction of pancreatic beta cells. SCOPE OF REVIEW In recent years, the role of immune checkpoints in the treatment of cancer has been increasingly recognized, but unfortunately, little attention has been paid to the significant role they play both in the development of secondary diabetes with immune checkpoint inhibitors and the treatment of T1D, such as cytotoxic T-lymphocyte antigen 4(CTLA-4), programmed cell death protein-1(PD-1), lymphocyte activation gene-3(LAG-3), programmed death ligand-1(PD-L1), and T-cell immunoglobulin mucin protein-3(TIM-3). Here, this review summarizes recent research on the role and mechanisms of diverse immune checkpoint molecules in mediating the development of T1D and their potential and theoretical basis for the prevention and treatment of diabetes. MAJOR CONCLUSIONS Immune checkpoint inhibitors related diabetes, similar to T1D, are severe endocrine toxicity induced with immune checkpoint inhibitors. Interestingly, numerous treatment measures show excellent efficacy for T1D via regulating diverse immune checkpoint molecules, including co-inhibitory and co-stimulatory molecules. Thus, targeting immune checkpoint molecules may exhibit potential for T1D treatment and improve clinical outcomes.
Collapse
Affiliation(s)
- Jia-Tong Ding
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical Medicine School, Nanchang University, Nanchang, China
| | - Kang-Ping Yang
- The Second Clinical Medicine School, Nanchang University, Nanchang, China
| | - Kong-Lan Lin
- The Second Clinical Medicine School, Nanchang University, Nanchang, China
| | - Yu-Ke Cao
- School of Ophthalmology & Optometry, Nanchang University, Nanchang, China
| | - Fang Zou
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Fang Zou,
| |
Collapse
|
17
|
Weigand K, Peschel G, Grimm J, Luu K, Schacherer D, Wiest R, Müller M, Schwarz H, Buechler C. Soluble CD137 is a novel serum marker of liver cirrhosis in patients with hepatitis C and alcohol-associated disease etiology. Eur J Immunol 2021; 52:633-645. [PMID: 34914098 DOI: 10.1002/eji.202149488] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/24/2021] [Accepted: 12/10/2021] [Indexed: 11/06/2022]
Abstract
Defective T-cell functions play a role in the persistence of HCV infection. Activated T cells express CD137, which costimulates antivirus T-cell responses, and this activity is antagonized by soluble CD137 (sCD137). Here, we show that in sera of 81 patients with chronic HCV, sCD137 levels did not correlate with measures of viral infection, and did not decline after virus eradication using direct-acting antivirals. Thus, serum sCD137 was similar in patients infected with HCV and in uninfected controls. Of note, in HCV patients with liver cirrhosis and patients with mostly alcohol-associated liver cirrhosis, sCD137 was increased. A negative association of sCD137 and albumin existed in both cohorts. sCD137 concentrations were similar in hepatic and portal vein blood excluding the liver as the origin of higher levels. Recombinant sCD137 reduced Th1 and Th2 but not Th17 cell polarization in vitro, and accordingly lowered IFN-γ, TNF, and IL-13 in cell media. Serum sCD137 is associated with inflammatory states, and positively correlated with serum TNF in cirrhotic HCV patients following virus eradication. Our study argues against a role of sCD137 in HCV infection and suggests a function of sCD137 in liver cirrhosis, which yet has to be defined.
Collapse
Affiliation(s)
- Kilian Weigand
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Georg Peschel
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Jonathan Grimm
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Khang Luu
- Department of Physiology and Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Doris Schacherer
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Reiner Wiest
- Department of Visceral Surgery and Medicine, University Inselspital, Bern, Switzerland
| | - Martina Müller
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Herbert Schwarz
- Department of Physiology and Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christa Buechler
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
18
|
Ajami M, Nazari M, mahmoodzadeh H, Moazzeni SM. Recombinant CD137-Fc, its synthesis, and applications for improving the immune system functions, such as tumor immunotherapy and to reduce the inflammation due to the novel coronavirus. J Cell Biochem 2021; 122:1072-1084. [PMID: 33993519 PMCID: PMC8242381 DOI: 10.1002/jcb.29928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/15/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022]
Abstract
CD137 (ILA/4-1BB), a member of tumor necrosis factor receptor superfamily, is one of the most important T cell costimulatory molecules. Interaction of this molecule with its ligand transmits a two-way signal that activates both T lymphocyte and antigen presenting cells. The soluble form of CD137 (sCD137) reduces the activity of its membrane isoform and is associated with T lymphocyte activation-induced cell death. Recombinant CD137-Fc may be used to treat cancers, autoimmune disorders and viral infections. It may also be useful for management of coronavirus infection. The 1276 bp DNA sequence encoded CD137-Fc recombinant protein was prepared and subcloned into lentiviral vector and expressed in transduced CHO-K1 eukaryotic cells. The sodium dodecyl sulfate-polyacrylamide gel electrophoresis, Western blot analysis, and enzyme-linked immunosorbent assay analysis results demonstrated that the expression of the 70-kDa CD137-Fc molecule was detectable without any degradation. This study helps to confirm previous research suggesting the use of this recombinant protein as a promising solution for the treatment of virus infections. CD137-Fc fusion protein could also make immunotherapy more effective for some diseases. This product is widely used in novel medical treatments, including cell-based immunotherapy such as dendritic cell, CAR T and CAR NK therapy. Its production and usage in research and treatment is noticeable also in current coronavirus disease 2019 pandemic.
Collapse
Affiliation(s)
- Maryam Ajami
- Department of Immunology, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Mahboobeh Nazari
- Monoclonal Antibody Research Center, Avicenna Research InstituteACECRTehranIran
| | | | | |
Collapse
|
19
|
Wong HY, Schwarz H. CD137 / CD137 ligand signalling regulates the immune balance: A potential target for novel immunotherapy of autoimmune diseases. J Autoimmun 2020; 112:102499. [PMID: 32505443 DOI: 10.1016/j.jaut.2020.102499] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 02/08/2023]
Abstract
CD137 (TNFRSF9, 4-1BB) is a potent co-stimulatory molecule of the tumour necrosis factor receptor superfamily (TNFRSF) that is expressed by activated T cells. CD137/CD137 ligand (CD137L) signalling primarily induces a potent cell-mediated immune response, while signalling of cell surface-expressed CD137L into antigen presenting cells enhances their activation, differentiation and migratory capacity. Studies have shown that bidirectional CD137/CD137L signalling plays an important role in the pathogenesis of autoimmune diseases. This review discusses the mechanisms how CD137/CD137L signalling contributes to immune deviation of helper T cell pathways in various murine models, and the potential of developing immunotherapies targeting CD137/CD137L signalling for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Hiu Yi Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117593, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456, Singapore
| | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117593, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456, Singapore.
| |
Collapse
|
20
|
Foda BM, Ciecko AE, Serreze DV, Ridgway WM, Geurts AM, Chen YG. The CD137 Ligand Is Important for Type 1 Diabetes Development but Dispensable for the Homeostasis of Disease-Suppressive CD137 + FOXP3 + Regulatory CD4 T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:2887-2899. [PMID: 32295876 PMCID: PMC7296588 DOI: 10.4049/jimmunol.1900485] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 03/31/2020] [Indexed: 01/25/2023]
Abstract
CD137 modulates type 1 diabetes (T1D) progression in NOD mice. We previously showed that CD137 expression in CD4 T cells inhibits T1D, but its expression in CD8 T cells promotes disease development by intrinsically enhancing the accumulation of β-cell-autoreactive CD8 T cells. CD137 is expressed on a subset of FOXP3+ regulatory CD4 T cells (Tregs), and CD137+ Tregs are the main source of soluble CD137. Soluble CD137 suppresses T cells in vitro by binding to the CD137 ligand (CD137L) upregulated on activated T cells. To further study how the opposing functions of CD137 are regulated, we successfully targeted Tnfsf9 (encoding CD137L) in NOD mice using the CRISPR/Cas9 system (designated NOD.Tnfsf9 -/-). Relative to wild-type NOD mice, T1D development in the NOD.Tnfsf9 -/- strain was significantly delayed, and mice developed less insulitis and had reduced frequencies of β-cell-autoreactive CD8 T cells. Bone marrow chimera experiments showed that CD137L-deficient hematopoietic cells were able to confer T1D resistance. Adoptive T cell transfer experiments showed that CD137L deficiency on myeloid APCs was associated with T1D suppression. Conversely, lack of CD137L on T cells enhanced their diabetogenic activity. Furthermore, neither CD137 nor CD137L was required for the development and homeostasis of FOXP3+ Tregs. However, CD137 was critical for the in vivo T1D-suppressive activity of FOXP3+ Tregs, suggesting that the interaction between CD137 and CD137L regulates their function. Collectively, our results provide new insights into the complex roles of CD137-CD137L interaction in T1D.
Collapse
Affiliation(s)
- Bardees M Foda
- Department of Molecular Genetics and Enzymology, National Research Centre, Dokki, 12622, Egypt
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226
- Max McGee National Research Center for Juvenile Diabetes, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Ashley E Ciecko
- Max McGee National Research Center for Juvenile Diabetes, Medical College of Wisconsin, Milwaukee, WI 53226
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226
| | | | - William M Ridgway
- Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, Davis, CA 95616
| | - Aron M Geurts
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226; and
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Yi-Guang Chen
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226;
- Max McGee National Research Center for Juvenile Diabetes, Medical College of Wisconsin, Milwaukee, WI 53226
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226
| |
Collapse
|