1
|
Abhiraman GC, Householder KD, Rodriguez GE, Glassman CR, Saxton RA, Breuer CB, Wilson SC, Su L, Yen M, Hsu C, Pillarisetty VG, Reticker-Flynn NE, Garcia KC. Redirecting immune signaling with cytokine adaptors. Nat Commun 2025; 16:2432. [PMID: 40069219 PMCID: PMC11897282 DOI: 10.1038/s41467-025-57681-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/28/2025] [Indexed: 03/15/2025] Open
Abstract
Cytokines are signaling molecules that coordinate complex immune processes and are frequently dysregulated in disease. While cytokine blockade has become a common therapeutic modality, cytokine agonism has had limited utility due to the widespread expression of cytokine receptors with pleiotropic effects. To overcome this limitation, we devise an approach to engineer molecular switches, termed cytokine adaptors, that transform one cytokine signal into an alternative signal with a different functional output. Endogenous cytokines act to nucleate the adaptors, converting the cytokine-adaptor complex into a surrogate agonist for a different cytokine pathway. In this way, cytokine adaptors, which have no intrinsic agonist activity, can function as conditional, context-dependent agonists. We develop cytokine adaptors that convert IL-10 or TGF-β into IL-2 receptor agonists to reverse T cell suppression. We also convert the pro-inflammatory cytokines IL-23 or IL-17 into immunosuppressive IL-10 receptor agonists. Thus, we show that cytokine adaptors can convert immunosuppressive cytokines into immunostimulatory cytokines, or vice versa. Unlike other methods of immune conversion that require cell engineering, cytokine adaptors are soluble molecules that leverage endogenous cues from the microenvironment to drive context-specific signaling.
Collapse
Affiliation(s)
- Gita C Abhiraman
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Karsten D Householder
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Grayson E Rodriguez
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Caleb R Glassman
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
| | - Robert A Saxton
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
| | - Cort B Breuer
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Steven C Wilson
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
| | - Leon Su
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
| | - Michelle Yen
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA
| | - Cynthia Hsu
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, 98112, USA
| | - Venu G Pillarisetty
- Department of Surgery, University of Washington School of Medicine, Seattle, WA, 98112, USA
| | - Nathan E Reticker-Flynn
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA, 94305, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
2
|
Cao W, Yang Z, Mo L, Liu Z, Wang J, Zhang Z, Wang K, Pan W. Causal relationship between immune cells and risk of heart failure: evidence from a Mendelian randomization study. Front Cardiovasc Med 2025; 11:1473905. [PMID: 39917605 PMCID: PMC11798955 DOI: 10.3389/fcvm.2024.1473905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/26/2024] [Indexed: 02/09/2025] Open
Abstract
Background Heart failure (HF) is a clinical syndrome resulting from structural damage or dysfunction of the heart. Previous investigations have highlighted the critical involvement of immune cells in the progression of heart failure, with distinct roles attributed to different types of immune cells. The objective of the current research was to explore the potential connections between immune characteristics and the development of HF, as well as to ascertain the nature of the causality between these factors. Methods To assess the causal association of immunological profiles with HF based on publicly available genome-wide studies, we employed a two-sample Mendelian randomization technique, utilizing the inverse variance weighted (IVW) method as our primary analytical approach. In addition, we assessed heterogeneity and cross-sectional pleiotropy through sensitivity analyses. Results A two-sample Mendelian randomization (MR) analysis was conducted using IVW as the primary method. At a significance level of 0.001, we identified 40 immunophenotypes that have a significant causal relationship with HF. There is a significant causal relationship between these phenotypes and heart failure. These immunophenotypes, 8 of which were in B cells, 5 in cDC, 2 in T cell maturation stage, 2 in monocytes, 3 in myeloid cells, 7 in TBNK and 13 in Treg. Sensitivity analyses were conducted to validate the strength and reliability of the MR findings. Conclusions Our study suggests that there appears to be a causal effect between multiple immune cells on heart failure. This discovery provides a new avenue for the development of therapeutic treatments for HF and a new target for drug development.
Collapse
Affiliation(s)
- Wenjing Cao
- Department of Cardiology, Department of Geriatric Medicine, Foshan Women and Children's Hospital, Foshan, Guangdong, China
| | - Zefu Yang
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, China
| | - Liumei Mo
- Department of Cardiology, Department of Geriatric Medicine, Foshan Women and Children's Hospital, Foshan, Guangdong, China
| | - Zhenhao Liu
- Department of Cardiovascular Medicine, Pingxiang People's Hospital, Jiangxi, China
| | - Jiawei Wang
- Department of Critical Care Medicine, Jieyang Third People's Hospital, Jieyang, Guangdong, China
| | - Zhenhong Zhang
- Department of Cardiology Medical, The Second People's Hospital of Foshan, Foshan, China
| | - Kui Wang
- The First Clinical Medical College, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wei Pan
- Department of Cardiology, Department of Geriatric Medicine, Foshan Women and Children's Hospital, Foshan, Guangdong, China
| |
Collapse
|
3
|
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
|
4
|
Alvarez-Salazar EK, Cortés-Hernández A, Arteaga-Cruz S, Soldevila G. Induced regulatory T cells as immunotherapy in allotransplantation and autoimmunity: challenges and opportunities. J Leukoc Biol 2024; 116:947-965. [PMID: 38630873 DOI: 10.1093/jleuko/qiae062] [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: 12/01/2023] [Revised: 02/14/2024] [Accepted: 02/21/2024] [Indexed: 04/19/2024] Open
Abstract
Regulatory T cells play a crucial role in the homeostasis of the immune response. Regulatory T cells are mainly generated in the thymus and are characterized by the expression of Foxp3, which is considered the regulatory T-cell master transcription factor. In addition, regulatory T cells can be induced from naive CD4+ T cells to express Foxp3 under specific conditions both in vivo (peripheral regulatory T cells) and in vitro (induced regulatory T cells). Both subsets of thymic regulatory T cells and peripheral regulatory T cells are necessary for the establishment of immune tolerance to self and non-self antigens. Although it has been postulated that induced regulatory T cells may be less stable compared to regulatory T cells, mainly due to epigenetic differences, accumulating evidence in animal models shows that induced regulatory T cells are stable in vivo and can be used for the treatment of inflammatory disorders, including autoimmune diseases and allogeneic transplant rejection. In this review, we describe the biological characteristics of induced regulatory T cells, as well as the key factors involved in induced regulatory T-cell transcriptional, metabolic, and epigenetic regulation, and discuss recent advances for de novo generation of stable regulatory T cells and their use as immunotherapeutic tools in different experimental models. Moreover, we discuss the challenges and considerations for the application of induced regulatory T cells in clinical trials and describe the new approaches proposed to achieve in vivo stability, including functional or metabolic reprogramming and epigenetic editing.
Collapse
Affiliation(s)
- Evelyn Katy Alvarez-Salazar
- Department of Immunology and National Laboratory of Flow Cytometry, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar s/n, Ciudad Universitaria, Colonia Copilco, Delegación Coyoacan, Apartado Postal 70228, CP 04510 Mexico City, Mexico
| | - Arimelek Cortés-Hernández
- Department of Immunology and National Laboratory of Flow Cytometry, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar s/n, Ciudad Universitaria, Colonia Copilco, Delegación Coyoacan, Apartado Postal 70228, CP 04510 Mexico City, Mexico
| | - Saúl Arteaga-Cruz
- Department of Immunology and National Laboratory of Flow Cytometry, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar s/n, Ciudad Universitaria, Colonia Copilco, Delegación Coyoacan, Apartado Postal 70228, CP 04510 Mexico City, Mexico
| | - Gloria Soldevila
- Department of Immunology and National Laboratory of Flow Cytometry, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar s/n, Ciudad Universitaria, Colonia Copilco, Delegación Coyoacan, Apartado Postal 70228, CP 04510 Mexico City, Mexico
| |
Collapse
|
5
|
Khalilollah S, Kalantari Soltanieh S, Obaid Saleh R, Ali Alzahrani A, Ghaleb Maabreh H, Mazin Al-Hamdani M, Dehghani-Ghorbi M, Shafiei Khonachaei M, Akhavan-Sigari R. LncRNAs involvement in pathogenesis of immune-related disease via regulation of T regulatory cells, an updated review. Cytokine 2024; 179:156585. [PMID: 38579428 DOI: 10.1016/j.cyto.2024.156585] [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: 05/24/2023] [Revised: 03/03/2024] [Accepted: 03/20/2024] [Indexed: 04/07/2024]
Abstract
The pathophysiology of several illnesses, including cancer and autoimmune diseasesdepends on human regulatory T cells (Tregs), and abnormalities in these cells may function as triggers for these conditions. Cancer and autoimmune, and gynecological diseases are associated with the differentiation of the proinflammatory T cell subset TH17 and its balance with the production of Treg. Recently, long non-coding RNAs (lncRNAs) have become important regulatory molecules in a wide range of illnesses. During epigenetic regulation, they can control the expression of important genes at several levels by affecting transcription, post-transcriptional actions, translation, and protein modification. They might connect with different molecules, such as proteins, DNA and RNA, and their structural composition is intricate. Because lncRNAs regulatebiological processes, including cell division, death, and growth, they are linked to severaldiseases. A notable instance of this is the lncRNA NEAT1, which has been the subject of several investigations to ascertain its function in immune cell development. In the context of immune cell development, several additional lncRNAs have been connected to Treg cell differentiation. In this work, we summarize current findings about the diverse functions of lncRNAs in Treg cell differentiation and control of the Th17/Treg homeostasis in autoimmune disorders, cancers, as well as several gynecological diseases where Tregs are key players.
Collapse
Affiliation(s)
- Shayan Khalilollah
- Department of Neurosurgery, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | | | - Raed Obaid Saleh
- Department of Pathological Analysis, College of Applied Science, University of Fallujah, Al-Anbar, Iraq.
| | | | - Hatem Ghaleb Maabreh
- Department of Dermatovenerology, Foreign Languages, RUDN University (Peoples' Friendship University of Russia named after Patrice Lumumba), Moscow, Russia.
| | | | - Mahmoud Dehghani-Ghorbi
- Hematology-Oncology Department, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Poland.
| |
Collapse
|
6
|
Ngai D, Sukka SR, Tabas I. Crosstalk between efferocytic myeloid cells and T-cells and its relevance to atherosclerosis. Front Immunol 2024; 15:1403150. [PMID: 38873597 PMCID: PMC11169609 DOI: 10.3389/fimmu.2024.1403150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024] Open
Abstract
The interplay between myeloid cells and T-lymphocytes is critical to the regulation of host defense and inflammation resolution. Dysregulation of this interaction can contribute to the development of chronic inflammatory diseases. Important among these diseases is atherosclerosis, which refers to focal lesions in the arterial intima driven by elevated apolipoprotein B-containing lipoproteins, notably low-density lipoprotein (LDL), and characterized by the formation of a plaque composed of inflammatory immune cells, a collection of dead cells and lipids called the necrotic core, and a fibrous cap. As the disease progresses, the necrotic core expands, and the fibrous cap becomes thin, which increases the risk of plaque rupture or erosion. Plaque rupture leads to a rapid thrombotic response that can give rise to heart attack, stroke, or sudden death. With marked lowering of circulating LDL, however, plaques become more stable and cardiac risk is lowered-a process known as atherosclerosis regression. A critical aspect of both atherosclerosis progression and regression is the crosstalk between innate (myeloid cells) and adaptive (T-lymphocytes) immune cells. Myeloid cells are specialized at clearing apoptotic cells by a process called efferocytosis, which is necessary for inflammation resolution. In advanced disease, efferocytosis is impaired, leading to secondary necrosis of apoptotic cells, inflammation, and, most importantly, defective tissue resolution. In regression, efferocytosis is reawakened aiding in inflammation resolution and plaque stabilization. Here, we will explore how efferocytosing myeloid cells could affect T-cell function and vice versa through antigen presentation, secreted factors, and cell-cell contacts and how this cellular crosstalk may contribute to the progression or regression of atherosclerosis.
Collapse
Affiliation(s)
- David Ngai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Santosh R. Sukka
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
- Department of Physiology, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
7
|
Su QY, Li HC, Jiang XJ, Jiang ZQ, Zhang Y, Zhang HY, Zhang SX. Exploring the therapeutic potential of regulatory T cell in rheumatoid arthritis: Insights into subsets, markers, and signaling pathways. Biomed Pharmacother 2024; 174:116440. [PMID: 38518605 DOI: 10.1016/j.biopha.2024.116440] [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: 01/21/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024] Open
Abstract
Rheumatoid arthritis (RA) is a complex autoimmune inflammatory rheumatic disease characterized by an imbalance between immunological reactivity and immune tolerance. Regulatory T cells (Tregs), which play a crucial role in controlling ongoing autoimmunity and maintaining peripheral tolerance, have shown great potential for the treatment of autoimmune inflammatory rheumatic diseases such as RA. This review aims to provide an updated summary of the latest insights into Treg-targeting techniques in RA. We focus on current therapeutic strategies for targeting Tregs based on discussing their subsets, surface markers, suppressive function, and signaling pathways in RA.
Collapse
Affiliation(s)
- Qin-Yi Su
- The Second Hospital of Shanxi Medical University, Department of Rheumatology, Taiyuan, China; Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Huan-Cheng Li
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Xiao-Jing Jiang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Zhong-Qing Jiang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Yan Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - He-Yi Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Sheng-Xiao Zhang
- The Second Hospital of Shanxi Medical University, Department of Rheumatology, Taiyuan, China; Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China.
| |
Collapse
|
8
|
Valizadeh M, Raoofian R, Homayoonfar A, Hajati E, Pourfathollah AA. MARCH-I: A negative regulator of dendritic cell maturation. Exp Cell Res 2024; 436:113946. [PMID: 38331309 DOI: 10.1016/j.yexcr.2024.113946] [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: 10/14/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/10/2024]
Abstract
The expression of costimulatory molecules such as MHC-II, CD86 and CD83 on dendritic cells (DCs) are strongly regulated during cellular activation. Ubiquitination of some of these markers by the E3 ubiquitin ligase MARCH-I affects the maturation state of DCs and subsequently modulates immune responses. The effects of MARCH-I gene overexpression on the functional activity of human DCs is not well understood. Here, we investigate how MARCH-I, regulates maturation of DCs. We now provide evidence that MARCH-I transduced DCs secrete high levels of IL10 despite low secretion of IL 6 and IL 12 in response to LPS stimulation. They are weak stimulators of T lymphocyte cells but skewed T cell polarization toward T regulatory subset. These results exhibit that reduced expression of surface costimulatory molecules suppresses DC activation. It can be concluded that overexpression of MARCH-I gene in DCs leads to the production of tolerogenic DC.
Collapse
Affiliation(s)
- Maryam Valizadeh
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza Raoofian
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Afrooz Homayoonfar
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Esmerdis Hajati
- Iranian Blood Transfusion Organization Research Center, Tehran, Iran
| | - Ali A Pourfathollah
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
9
|
Darvish Z, Kheder RK, Faraj TA, Najmaldin SK, Mollazadeh S, Nosratabadi R, Esmaeili SA. A better understanding of the role of the CTLA-CD80/86 axis in the treatment of autoimmune diseases. Cell Biochem Funct 2024; 42:e3895. [PMID: 38050849 DOI: 10.1002/cbf.3895] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/10/2023] [Accepted: 11/17/2023] [Indexed: 12/07/2023]
Abstract
Autoimmune diseases are diseases in which the regulatory mechanisms of the immune response are disturbed. As a result, the body loses self-tolerance. Since one of the main regulatory mechanisms of the immune response is the CTLA4-CD80/86 axis, this hypothesis suggests that autoimmune diseases potentially share a similar molecular basis of pathogenesis. Hence, investigating the CTLA4-CD80/86 axis may be helpful in finding an appropriate treatment strategy. Therefore, this study aims to investigate the molecular basis of the CTLA4-CD80/86 axis in the regulation of the immune response, and then its role in developing some autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, type 1 diabetes, and multiple sclerosis. As well, the main therapeutic strategies affecting the CTLA4-CD80/86 axis have been summarized to highlight the importance of this axis in management of autoimmune diseases.
Collapse
Affiliation(s)
- Zahra Darvish
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramiar Kamal Kheder
- Medical Laboratory Science Department, College of Science, University of Raparin, Rania, Sulaymaniyah, Iraq
| | - Tola Abdulsattar Faraj
- Department of Basic Sciences, College of Medicine, Hawler Medical University, Erbil, Iraq
| | - Soran K Najmaldin
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center٫ North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Reza Nosratabadi
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Gastroenterology and Hepatology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Coronel PMV, Pereira IC, Basilio DCLS, Espinoça IT, de Souza KFS, Ota RSN, de Almeida EB, Paredes-Gamero EJ, Wilhelm Filho D, Perdomo RT, Parisotto EB. Biomarkers of oxidative stress and inflammation in subjects with COVID-19: Characterization and prognosis of the disease. Microb Pathog 2023; 184:106339. [PMID: 37690769 DOI: 10.1016/j.micpath.2023.106339] [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: 06/01/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023]
Abstract
Coronavirus disease (COVID-19) is an acute respiratory disease caused by the new coronavirus (SARS-CoV-2) that has spread throughout the world causing millions of deaths. COVID-19 promotes excessive release of pro-inflammatory cytokines leading to acute lung injury and death. Reactive oxygen species (ROS) and oxidative stress (OS) may also play a role in the pathophysiology of COVID-19. The present study investigated levels of inflammatory cytokines (IL-1β, IL-6, IL-8, IL-10, IL-12) and OS biomarkers (MPO, SOD, CAT, GST enzymes and contents of GSH, TBARS and PC) in patients with SARS-CoV-2 infection, which were correlated with disease severity. Patients with SARS significantly increased IL-1β levels, while IL-6 levels were elevated in both groups of SARS-CoV-2 positive patients. The most severe patients showed increased levels of IL-8 and IL-10, while subjects without SARS showed lower values. MPO activity were higher in both groups of SARS-CoV-2 positive patients, while SOD and CAT activity were decreased in both groups. Compared to controls, GGT was elevated only in the SARS patient group, while GST values were increased in the group of positive patients in SARS-CoV-2 without SARS and were decreased in patients with SARS. GSH and UA contents decreased in SARS-CoV-2 positive subjects, whereas TBARS and PC contents increased in both groups of SARS-CoV-2 positive patients, particularly in the SARS patient group. In addition, several important correlations were found between cytokines and the different OS parameters suggesting some inter-relationship in the complex antioxidant system of the patients. In general, patients with SARS-CoV-2 infection showed higher levels of OS biomarkers, and also elevated contents of IL-6 and IL-10, probably worsening the damage caused by SARS-CoV-2 infection. This damage may contribute to the severity of the disease and its complications, as well as a prognosis for SARS-CoV-2 patients.
Collapse
Affiliation(s)
- Paola Mayara Valente Coronel
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso Do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Indiara Correia Pereira
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso Do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Denise Caroline Luiz Soares Basilio
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso Do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Isabelly Teixeira Espinoça
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso Do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | | | - Rafael Seiji Nakano Ota
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso Do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | | | - Edgar Julian Paredes-Gamero
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso Do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Danilo Wilhelm Filho
- Departamento de Ecologia e Zoologia, Centro de Ciências Biológicas (CCB), Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Renata Trentin Perdomo
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso Do Sul, Campo Grande, Mato Grosso do Sul, Brazil.
| | - Eduardo Benedetti Parisotto
- Faculdade de Ciências Farmacêuticas Alimentos e Nutrição (FACFAN), Universidade Federal de Mato Grosso Do Sul, Campo Grande, Mato Grosso do Sul, Brazil.
| |
Collapse
|
11
|
Fruntealată RF, Marius M, Boboc IKS, Mitran SI, Ciurea ME, Stoica GA. Mechanisms of Altered Immune Response in Skin Melanoma. CURRENT HEALTH SCIENCES JOURNAL 2023; 49:297-311. [PMID: 38314217 PMCID: PMC10832881 DOI: 10.12865/chsj.49.03.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/18/2023] [Indexed: 02/06/2024]
Abstract
Melanoma, a deadly form of skin cancer, poses significant challenges to the host immune system, allowing tumor cells to evade immune surveillance and persist. This complex interplay between melanoma and the immune system involves a multitude of mechanisms that impair immune recognition and promote tumor progression. This review summarizes the intricate strategies employed by melanoma cells to evade the immune response, including defective immune recognition, immune checkpoint activation, and the role of regulatory T-cells, myeloid-derived suppressor cells, and exosomes in suppressing anti-tumor immunity. Additionally, we discuss potential therapeutic targets aimed at reversing immune evasion in melanoma, highlighting the importance of understanding these mechanisms for developing more effective immunotherapies. Improved insights into the interactions between melanoma and the immune system will aid in the development of novel treatment strategies to enhance anti-tumor immune responses and improve patient outcomes.
Collapse
Affiliation(s)
| | - Matei Marius
- Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
| | - Ianis Kevyn Stefan Boboc
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Romania
| | | | - Marius Eugen Ciurea
- Department of Physiology, University of Medicine and Pharmacy of Craiova, Romania
| | - George-Alin Stoica
- Department of Pediatric Surgery, University of Medicine and Pharmacy of Craiova, Romania
| |
Collapse
|
12
|
Avila JP, Carvalho BM, Coimbra EC. A Comprehensive View of the Cancer-Immunity Cycle (CIC) in HPV-Mediated Cervical Cancer and Prospects for Emerging Therapeutic Opportunities. Cancers (Basel) 2023; 15:1333. [PMID: 36831674 PMCID: PMC9954575 DOI: 10.3390/cancers15041333] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Cervical cancer (CC) is the fourth most common cancer in women worldwide, with more than 500,000 new cases each year and a mortality rate of around 55%. Over 80% of these deaths occur in developing countries. The most important risk factor for CC is persistent infection by a sexually transmitted virus, the human papillomavirus (HPV). Conventional treatments to eradicate this type of cancer are accompanied by high rates of resistance and a large number of side effects. Hence, it is crucial to devise novel effective therapeutic strategies. In recent years, an increasing number of studies have aimed to develop immunotherapeutic methods for treating cancer. However, these strategies have not proven to be effective enough to combat CC. This means there is a need to investigate immune molecular targets. An adaptive immune response against cancer has been described in seven key stages or steps defined as the cancer-immunity cycle (CIC). The CIC begins with the release of antigens by tumor cells and ends with their destruction by cytotoxic T-cells. In this paper, we discuss several molecular alterations found in each stage of the CIC of CC. In addition, we analyze the evidence discovered, the molecular mechanisms and their relationship with variables such as histological subtype and HPV infection, as well as their potential impact for adopting novel immunotherapeutic approaches.
Collapse
Affiliation(s)
| | | | - Eliane Campos Coimbra
- Institute of Biological Sciences, University of Pernambuco (ICB/UPE), Rua Arnóbio Marques, 310, Santo Amaro, Recife 50100-130, PE, Brazil
| |
Collapse
|
13
|
Li R, Li H, Yang X, Hu H, Liu P, Liu H. Crosstalk between dendritic cells and regulatory T cells: Protective effect and therapeutic potential in multiple sclerosis. Front Immunol 2022; 13:970508. [PMID: 36177043 PMCID: PMC9513370 DOI: 10.3389/fimmu.2022.970508] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system related to autoimmunity and is characterized by demyelination, neuroinflammation, and neurodegeneration. Cell therapies mediated by dendritic cells (DCs) and regulatory T cells (Tregs) have gradually become accumulating focusing in MS, and the protective crosstalk mechanisms between DCs and Tregs provide the basis for the efficacy of treatment regimens. In MS and its animal model experimental autoimmune encephalomyelitis, DCs communicate with Tregs to form immune synapses and complete a variety of complex interactions to counteract the unbalanced immune tolerance. Through different co-stimulatory/inhibitory molecules, cytokines, and metabolic enzymes, DCs regulate the proliferation, differentiation and function of Tregs. On the other hand, Tregs inhibit the mature state and antigen presentation ability of DCs, ultimately improving immune tolerance. In this review, we summarized the pivotal immune targets in the interaction between DCs and Tregs, and elucidated the protective mechanisms of DC-Treg cell crosstalk in MS, finally interpreted the complex cell interplay in the manner of inhibitory feedback loops to explore novel therapeutic directions for MS.
Collapse
Affiliation(s)
- Ruoyu Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyan Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huiru Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peidong Liu
- Department of Neurosurgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Translational Medicine Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongbo Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Translational Medicine Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Hongbo Liu,
| |
Collapse
|
14
|
Shiri Aghbash P, Shirvaliloo M, Khalo Abass Kasho A, Alinezhad F, Nauwynck H, Bannazadeh Baghi H. Cluster of differentiation frequency on antigen presenting-cells: The next step to cervical cancer prognosis? Int Immunopharmacol 2022; 108:108896. [DOI: 10.1016/j.intimp.2022.108896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/16/2022] [Accepted: 05/23/2022] [Indexed: 11/29/2022]
|
15
|
Li F, Long Y, Yu X, Tong Y, Gong L. Different Immunoregulation Roles of Activin A Compared With TGF-β. Front Immunol 2022; 13:921366. [PMID: 35774793 PMCID: PMC9237220 DOI: 10.3389/fimmu.2022.921366] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Activin A, a critical member of the transforming growth factor-β (TGF-β) superfamily, is a pluripotent factor involved in allergies, autoimmune diseases, cancers and other diseases with immune disorder. Similar to its family member, TGF-β, activin A also transmits signals through SMAD2/SMAD3, however, they bind to distinct receptors. Recent studies have uncovered that activin A plays a pivotal role in both innate and adaptive immune systems. Here we mainly focus its effects on activation, differentiation, proliferation and function of cells which are indispensable in the immune system and meanwhile make some comparisons with those of TGF-β.
Collapse
Affiliation(s)
- Fanglin Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yiru Long
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaolu Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yongliang Tong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Likun Gong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- *Correspondence: Likun Gong,
| |
Collapse
|
16
|
Lu Y, Xia N, Cheng X. Regulatory T Cells in Chronic Heart Failure. Front Immunol 2021; 12:732794. [PMID: 34630414 PMCID: PMC8493934 DOI: 10.3389/fimmu.2021.732794] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/02/2021] [Indexed: 12/21/2022] Open
Abstract
Heart failure is a global problem with high hospitalization and mortality rates. Inflammation and immune dysfunction are involved in this disease. Owing to their unique function, regulatory T cells (Tregs) have reacquired attention recently. They participate in immunoregulation and tissue repair in the pathophysiology of heart failure. Tregs are beneficial in heart by suppressing excessive inflammatory responses and promoting stable scar formation in the early stage of heart injury. However, in chronic heart failure, the phenotypes and functions of Tregs changed. They transformed into an antiangiogenic and profibrotic cell type. In this review, we summarized the functions of Tregs in the development of chronic heart failure first. Then, we focused on the interactions between Tregs and their target cells. The target cells of Tregs include immune cells (such as monocytes/macrophages, dendritic cells, T cells, and B cells) and parenchymal cells (such as cardiomyocytes, fibroblasts, and endothelial cells). Next-generation sequencing and gene editing technology make immunotherapy of heart failure possible. So, prospective therapeutic approaches based on Tregs in chronic heart failure had also been evaluated.
Collapse
Affiliation(s)
- Yuzhi Lu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ni Xia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Iida Y, Yoshikawa R, Murata A, Kotani H, Kazuki Y, Oshimura M, Matsuzaki Y, Harada M. Local injection of CCL19-expressing mesenchymal stem cells augments the therapeutic efficacy of anti-PD-L1 antibody by promoting infiltration of immune cells. J Immunother Cancer 2021; 8:jitc-2020-000582. [PMID: 32675195 PMCID: PMC7368496 DOI: 10.1136/jitc-2020-000582] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2020] [Indexed: 12/12/2022] Open
Abstract
Background Mesenchymal stem/stromal cells (MSC) accumulate and reside in tumor sites. Methods Taking advantage of this feature in anticancer therapy, immortalized murine MSC (iMSC) were genetically altered to produce chemokine (C-C motif) ligand 19 (iMSC/CCL19), which attracts dendritic cells (DC) and T lymphocytes. Thereafter, iMSC/CCL19 were examined for their therapeutic efficacy using a syngeneic CT26 colon carcinoma cell line. Results Co-injection of iMSC/CCL19 into mice significantly suppressed the in vivo growth of CT26 cells compared with that of CCL19-expressing immortalized fibroblasts (iFib/CCL19). This anticancer effect was not observed when injected in CT26-bearing nude mice. Co-injected iMSC/CCL19 survived longer than iFib/CCL19 in the tumor sites. In a therapeutic model, local injection of iMSC/CCL19 suppressed the tumor growth, and increased IFN (interferon)-γ+ CD8+ T cells and CCR7+ DC infiltration in tumor site was observed when treated with iMSC/CCL19, but not with iMSC. This antitumor effect was completely negated by depletion of CD4+ cells and partially negated by depletion of CD8+ cells. Furthermore, the antitumor effects induced by local injection of iMSC/CCL19 were augmented by additional therapy with anti-programmed death (PD)-ligand 1 (PD-L1) antibody, but not with anti-PD-1 antibody. This combination therapy cured most of the tumors in CT26-bearing mice. Conclusion These results suggest that local therapy with iMSC/CCL19 can suppress tumor growth via effective recruitment of CCR7+ DC into tumor sites and increase IFN-γ+ CD8+ T cells, and that combination with anti-PD-L1 antibody therapy can be a powerful anticancer therapy.
Collapse
Affiliation(s)
- Yuichi Iida
- Immunology, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Japan
| | - Rintaro Yoshikawa
- Life Science, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Shimane, Japan
| | - Akihiko Murata
- Molecular and Cellular Biology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Hitoshi Kotani
- Immunology, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Japan
| | - Yasuhiro Kazuki
- Biomedical Science, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Mitsuo Oshimura
- Chromosome Engineering Research Center, Tottori University, Yonago, Tottori, Japan
| | - Yumi Matsuzaki
- Life Science, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Shimane, Japan
| | - Mamoru Harada
- Immunology, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Japan
| |
Collapse
|
18
|
Li Q, Li X, Quan H, Wang Y, Qu G, Shen Z, He C. IL-10 -/- Enhances DCs Immunity Against Chlamydia psittaci Infection via OX40L/NLRP3 and IDO/Treg Pathways. Front Immunol 2021; 12:645653. [PMID: 34093535 PMCID: PMC8176032 DOI: 10.3389/fimmu.2021.645653] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 05/04/2021] [Indexed: 12/30/2022] Open
Abstract
Chlamydia psittaci (C. psittaci) is a common zoonotic agent that affects both poultry and humans. Interleukin 10 (IL-10) is an anti-inflammatory factor produced during chlamydial infection, while dendritic cells (DCs) are powerful antigen-presenting cells that induce a primary immune response in the host. However, IL-10 and DCs regulatory mechanisms in C. psittaci infection remain elusive. In vivo and in vitro investigations of the regulatory mechanisms were performed. IL-10−/− mice, conditional DCs depletion mice (zinc finger dendritic cell-diphtheria toxin receptor [zDC-DTR]), and double-deficient mice (DD, IL-10−/−/zDCDTR/DTR) were intranasally infected with C. psittaci. The results showed that more than 90% of IL-10−/− mice, 70% of wild-type mice, and 60% of double-deficient mice survived, whereas all zDC-DTR mice died. A higher lymphocyte proliferation index was found in the IL-10 inhibitor mice and IL-10−/− mice. Moreover, severe lesions and high bacterial loads were detected in the zDC-DTR mice compared with double-deficient mice. In vitro studies revealed increased OX40-OX40 ligand (OX40-OX40L) activation and CD4+T cell proliferation. Besides, the expression of indoleamine 2, 3-dioxygenase (IDO), and regulatory T cells were significantly reduced in the co-culture system of CD4+ T cells and IL-10−/− DCs in C. psittaci infection. Additionally, the activation of the NLR family pyrin domain-containing 3 (NLRP3) inflammasome increased to facilitate the apoptosis of DCs, leading to rapid clearance of C. psittaci. Our study showed that IL-10−/− upregulated the function of deficient DCs by activating OX40-OX40L, T cells, and the NLPR3 inflammasome, and inhibiting IDO, and regulatory T cells. These effects enhanced the survival rate of mice and C. psittaci clearance. Our research highlights the mechanism of IL-10 interaction with DCs, OX40-OX40L, and the NLPR3 inflammasome, as potential targets against C. psittaci infection.
Collapse
Affiliation(s)
- Qiang Li
- Key Lab of Animal Epidemiology and Zoonoses of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaohui Li
- Key Lab of Animal Epidemiology and Zoonoses of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hongkun Quan
- Key Lab of Animal Epidemiology and Zoonoses of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yihui Wang
- Key Lab of Animal Epidemiology and Zoonoses of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Guanggang Qu
- Preventive Veterinary Research Group, Binzhou Animal Science and Veterinary Medicine Academy of Shandong Province, Binzhou, China
| | - Zhiqiang Shen
- Preventive Veterinary Research Group, Binzhou Animal Science and Veterinary Medicine Academy of Shandong Province, Binzhou, China
| | - Cheng He
- Key Lab of Animal Epidemiology and Zoonoses of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
19
|
Recent Advancements in the Mechanisms Underlying Resistance to PD-1/PD-L1 Blockade Immunotherapy. Cancers (Basel) 2021; 13:cancers13040663. [PMID: 33562324 PMCID: PMC7915065 DOI: 10.3390/cancers13040663] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/03/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Immune checkpoint blockade targeting PD-1/PD-L1 has a promising therapeutic efficacy in different tumors, but a significant percentage of patients cannot benefit from this therapy due to primary and acquired resistance during treatment. This review summarizes the recent findings of PD-L1 role in resistance to therapies through the PD-1/PD-L1 pathway and other correlating signaling pathways. A special focus will be given to the key mechanisms underlying resistance to the PD-1/PD-L1 blockade in cancer immunotherapy. Furthermore, we also discuss the promising combination of therapeutic strategies for patients resistant to the PD-1/PD-L1 blockade in order to enhance the efficacy of immune checkpoint inhibitors. Abstract Release of immunoreactive negative regulatory factors such as immune checkpoint limits antitumor responses. PD-L1 as a significant immunosuppressive factor has been involved in resistance to therapies such as chemotherapy and target therapy in various cancers. Via interacting with PD-1, PD-L1 can regulate other factors or lead to immune evasion of cancer cells. Besides, immune checkpoint blockade targeting PD-1/PD-L1 has promising therapeutic efficacy in the different tumors, but a significant percentage of patients cannot benefit from this therapy due to primary and acquired resistance during treatment. In this review, we described the utility of PD-L1 expression levels for predicting poor prognosis in some tumors and present evidence for a role of PD-L1 in resistance to therapies through PD-1/PD-L1 pathway and other correlating signaling pathways. Afterwards, we elaborate the key mechanisms underlying resistance to PD-1/PD-L1 blockade in cancer immunotherapy. Furthermore, promising combination of therapeutic strategies for patients resistant to PD-1/PD-L1 blockade therapy or other therapies associated with PD-L1 expression was also summarized.
Collapse
|
20
|
Chen J, Zhang H, Zhou L, Hu Y, Li M, He Y, Li Y. Enhancing the Efficacy of Tumor Vaccines Based on Immune Evasion Mechanisms. Front Oncol 2021; 10:584367. [PMID: 33614478 PMCID: PMC7886973 DOI: 10.3389/fonc.2020.584367] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Tumor vaccines aim to expand tumor-specific T cells and reactivate existing tumor-specific T cells that are in a dormant or unresponsive state. As such, there is growing interest in improving the durable anti-tumor activity of tumor vaccines. Failure of vaccine-activated T cells to protect against tumors is thought to be the result of the immune escape mechanisms of tumor cells and the intricate immunosuppressive tumor microenvironment. In this review, we discuss how tumor cells and the tumor microenvironment influence the effects of tumor infiltrating lymphocytes and summarize how to improve the efficacy of tumor vaccines by improving the design of current tumor vaccines and combining tumor vaccines with other therapies, such as metabolic therapy, immune checkpoint blockade immunotherapy and epigenetic therapy.
Collapse
Affiliation(s)
- Jianyu Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Honghao Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lijuan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuxing Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Meifang Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yanjie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
21
|
Liu J, Cheng Y, Zheng M, Yuan B, Wang Z, Li X, Yin J, Ye M, Song Y. Targeting the ubiquitination/deubiquitination process to regulate immune checkpoint pathways. Signal Transduct Target Ther 2021; 6:28. [PMID: 33479196 PMCID: PMC7819986 DOI: 10.1038/s41392-020-00418-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/13/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022] Open
Abstract
The immune system initiates robust immune responses to defend against invading pathogens or tumor cells and protect the body from damage, thus acting as a fortress of the body. However, excessive responses cause detrimental effects, such as inflammation and autoimmune diseases. To balance the immune responses and maintain immune homeostasis, there are immune checkpoints to terminate overwhelmed immune responses. Pathogens and tumor cells can also exploit immune checkpoint pathways to suppress immune responses, thus escaping immune surveillance. As a consequence, therapeutic antibodies that target immune checkpoints have made great breakthroughs, in particular for cancer treatment. While the overall efficacy of immune checkpoint blockade (ICB) is unsatisfactory since only a small group of patients benefited from ICB treatment. Hence, there is a strong need to search for other targets that improve the efficacy of ICB. Ubiquitination is a highly conserved process which participates in numerous biological activities, including innate and adaptive immunity. A growing body of evidence emphasizes the importance of ubiquitination and its reverse process, deubiquitination, on the regulation of immune responses, providing the rational of simultaneous targeting of immune checkpoints and ubiquitination/deubiquitination pathways to enhance the therapeutic efficacy. Our review will summarize the latest findings of ubiquitination/deubiquitination pathways for anti-tumor immunity, and discuss therapeutic significance of targeting ubiquitination/deubiquitination pathways in the future of immunotherapy.
Collapse
Affiliation(s)
- Jiaxin Liu
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, 210002, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University School of Medicine, 210002, Nanjing, China
| | - Yicheng Cheng
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Ming Zheng
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, 210002, Nanjing, Jiangsu, China
| | - Bingxiao Yuan
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, 210002, Nanjing, Jiangsu, China
| | - Zimu Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, 210002, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University School of Medicine, 210002, Nanjing, China
| | - Xinying Li
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, 210002, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University School of Medicine, 210002, Nanjing, China
| | - Jie Yin
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University School of Medicine, 210002, Nanjing, China.
| | - Mingxiang Ye
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University School of Medicine, 210002, Nanjing, China.
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University School of Medicine, 210002, Nanjing, China.
| |
Collapse
|
22
|
Eddy K, Chen S. Overcoming Immune Evasion in Melanoma. Int J Mol Sci 2020; 21:E8984. [PMID: 33256089 PMCID: PMC7730443 DOI: 10.3390/ijms21238984] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/17/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023] Open
Abstract
Melanoma is the most aggressive and dangerous form of skin cancer that develops from transformed melanocytes. It is crucial to identify melanoma at its early stages, in situ, as it is "curable" at this stage. However, after metastasis, it is difficult to treat and the five-year survival is only 25%. In recent years, a better understanding of the etiology of melanoma and its progression has made it possible for the development of targeted therapeutics, such as vemurafenib and immunotherapies, to treat advanced melanomas. In this review, we focus on the molecular mechanisms that mediate melanoma development and progression, with a special focus on the immune evasion strategies utilized by melanomas, to evade host immune surveillances. The proposed mechanism of action and the roles of immunotherapeutic agents, ipilimumab, nivolumab, pembrolizumab, and atezolizumab, adoptive T- cell therapy plus T-VEC in the treatment of advanced melanoma are discussed. In this review, we implore that a better understanding of the steps that mediate melanoma onset and progression, immune evasion strategies exploited by these tumor cells, and the identification of biomarkers to predict treatment response are critical in the design of improved strategies to improve clinical outcomes for patients with this deadly disease.
Collapse
Affiliation(s)
- Kevinn Eddy
- Graduate Program in Cellular and Molecular Pharmacology, School of Graduate Studies Rutgers University, Piscataway, NJ 08854, USA;
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA
| | - Suzie Chen
- Graduate Program in Cellular and Molecular Pharmacology, School of Graduate Studies Rutgers University, Piscataway, NJ 08854, USA;
- Susan Lehman Cullman Laboratory for Cancer Research, Rutgers University, Piscataway, NJ 08854, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- Environmental & Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
23
|
Zhu B, Zhu L, Xia L, Xiong Y, Yin Q, Rui K. Roles of Ubiquitination and Deubiquitination in Regulating Dendritic Cell Maturation and Function. Front Immunol 2020; 11:586613. [PMID: 33329564 PMCID: PMC7717991 DOI: 10.3389/fimmu.2020.586613] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that play a key role in immune homeostasis and the adaptive immune response. DC-induced immune tolerance or activation is strictly dependent on the distinct maturation stages and migration ability of DCs. Ubiquitination is a reversible protein post-translational modification process that has emerged as a crucial mechanism that regulates DC maturation and function. Recent studies have shown that ubiquitin enzymes, including E3 ubiquitin ligases and deubiquitinases (DUBs), are pivotal regulators of DC-mediated immune function and serve as potential targets for DC-based immunotherapy of immune-related disorders (e.g., autoimmune disease, infections, and tumors). In this review, we summarize the recent progress regarding the molecular mechanisms and function of ubiquitination in DC-mediated immune homeostasis and immune response.
Collapse
Affiliation(s)
- Bo Zhu
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Zhu
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lin Xia
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China.,International Genome Center, Jiangsu University, Zhenjiang, China
| | - Yuyun Xiong
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Qing Yin
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ke Rui
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
24
|
Wang N, Yang L, Dai J, Wu Y, Zhang R, Jia X, Liu C. 5-FU inhibits migration and invasion of CRC cells through PI3K/AKT pathway regulated by MARCH1. Cell Biol Int 2020; 45:368-381. [PMID: 33085122 PMCID: PMC7894547 DOI: 10.1002/cbin.11493] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 12/11/2022]
Abstract
Colorectal cancer is a major health problem with a significant impact on the patients' quality of life. 5‐Fluorouracil is the most common chemotherapy drug used for this type of cancer. While its molecular mechanism is the inhibition of DNA synthesis via the inhibition of thymine nucleotide synthetase, its complete anticancer mechanism is not clear. Membrane‐associated RING‐CH‐1 (MARCH1) is an E3 ubiquitin ligase that plays an important role in antigen presentation. However, MARCH1 has not been studied in the context of colorectal cancer. In this study, we demonstrated that MARCH1 is highly expressed in colorectal cancer tissues and cell lines. Furthermore, migration and invasion of colorectal tumor cells were inhibited via transfection with small interfering RNAs to suppress the expression of MARCH1. The western blot analysis showed that MARCH1 regulates epithelial–mesenchymal transition and the PI3K/AKT pathway. Moreover, 5‐fluorouracil inhibited the proliferation, migration, and invasion of tumor cells, via the targeting of MARCH1 and the consequent downregulation of the PI3K/AKT pathway, impacting the progression of epithelial–mesenchymal transition. In conclusion, our study shows that MARCH1 may play a role as an oncogene in colorectal cancer and may represent a new target molecule of 5‐fluorouracil.
Collapse
Affiliation(s)
- Nuan Wang
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| | - Lijuan Yang
- Cancer Research Laboratory, Binzhou Medical University Hospital, Binzhou, China
| | - Juanjuan Dai
- Cancer Research Laboratory, Binzhou Medical University Hospital, Binzhou, China
| | - Yan Wu
- Cancer Research Laboratory, Binzhou Medical University Hospital, Binzhou, China
| | - Ranran Zhang
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| | - Xingfang Jia
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| | - Chengxia Liu
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
25
|
Akkaya B, Shevach EM. Regulatory T cells: Master thieves of the immune system. Cell Immunol 2020; 355:104160. [PMID: 32711171 DOI: 10.1016/j.cellimm.2020.104160] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/05/2020] [Accepted: 07/06/2020] [Indexed: 12/21/2022]
Abstract
Treg cells are the immune system's in-house combatants against pathological immune activation. Because they are vital to maintenance of peripheral tolerance, it is important to understand how they perform their functions. To this end, various mechanisms have been proposed for Treg-mediated immune inhibition. A major group of mechanisms picture Treg cells as skilled thieves stealing a plethora of molecules that would otherwise promote immune effector functions. This suggests that several million years of evolution have endowed Treg cells with efficient ways to deprive immune effectors of activating stimuli to prevent immunopathology for survival of the host. Although we are still long way from deciphering their complete set of tricks, this review will focus on the types of "crimes" committed by these master thieves in both secondary lymphoid organs and non-lymphoid tissue.
Collapse
Affiliation(s)
- Billur Akkaya
- Laboratory of Immune System Biology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ethan M Shevach
- Laboratory of Immune System Biology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
26
|
Tran GT, Hodgkinson SJ, Carter N, Verma ND, Robinson CM, Plain KM, Nomura M, Hall BM. Autoantigen specific IL-2 activated CD4 +CD25 +T regulatory cells inhibit induction of experimental autoimmune neuritis. J Neuroimmunol 2020; 341:577186. [PMID: 32058174 DOI: 10.1016/j.jneuroim.2020.577186] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/22/2020] [Accepted: 02/02/2020] [Indexed: 01/04/2023]
Abstract
Experimental autoimmune neuritis (EAN) induced by peripheral nerve myelin (PNM) is self-limiting and re-immunization with PNM does not re-activate disease. This study showed inhibition of EAN by CD4+CD25+T cells both from sensitized hosts or from naïve hosts after ex-vivo activation by PNM and rIL-2. Transfer of naïve CD4+CD25+T cells has no effect on EAN, nor did naïve CD4+CD25+T cells activated with rIL-2 and renal tubular antigen. Culture of naive CD4+CD25+Treg with rIL-2 and PNM induced mRNA for the IFN-gamma receptor. We showed naïve CD4+CD25+T cells activated by specific auto-antigen and rIL-2 produced more potent antigen-specific Treg that may have therapeutic potential.
Collapse
Affiliation(s)
- Giang T Tran
- Immune Tolerance Laboratory, Faculty of Medicine, UNSW Sydney, Ingham Institute, Liverpool, NSW, Australia.
| | - Suzanne J Hodgkinson
- Immune Tolerance Laboratory, Faculty of Medicine, UNSW Sydney, Ingham Institute, Liverpool, NSW, Australia; Departments of Neurology Liverpool Health Service, Liverpool, NSW, Australia.
| | - Nicole Carter
- Immune Tolerance Laboratory, Faculty of Medicine, UNSW Sydney, Ingham Institute, Liverpool, NSW, Australia.
| | - Nirupama D Verma
- Immune Tolerance Laboratory, Faculty of Medicine, UNSW Sydney, Ingham Institute, Liverpool, NSW, Australia.
| | - Catherine M Robinson
- Immune Tolerance Laboratory, Faculty of Medicine, UNSW Sydney, Ingham Institute, Liverpool, NSW, Australia.
| | - Karren M Plain
- Immune Tolerance Laboratory, Faculty of Medicine, UNSW Sydney, Ingham Institute, Liverpool, NSW, Australia.
| | - Masaru Nomura
- Immune Tolerance Laboratory, Faculty of Medicine, UNSW Sydney, Ingham Institute, Liverpool, NSW, Australia
| | - Bruce M Hall
- Immune Tolerance Laboratory, Faculty of Medicine, UNSW Sydney, Ingham Institute, Liverpool, NSW, Australia; Department of Nephrology, Liverpool Health Service, Liverpool, NSW, Australia.
| |
Collapse
|
27
|
Shangguan A, Shang N, Figini M, Pan L, Yang J, Ma Q, Hu S, Eresen A, Sun C, Wang B, Velichko Y, Yaghmai V, Zhang Z. Prophylactic dendritic cell vaccination controls pancreatic cancer growth in a mouse model. Cytotherapy 2020; 22:6-15. [PMID: 32005355 DOI: 10.1016/j.jcyt.2019.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 11/29/2019] [Accepted: 12/01/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related deaths with high recurrence after surgery due to a paucity of effective post-surgical adjuvant treatments. DC vaccines can activate multiple anti-tumor immune responses but have not been explored for post-surgery PDAC recurrence. Intraperitoneal (IP) delivery may allow increased DC vaccine dosage and migration to lymph nodes. Here, we investigated the role of prophylactic DC vaccination controlling PDAC tumor growth with IP delivery as an administration route for DC vaccination. METHODS DC vaccines were generated using ex vivo differentiation and maturation of bone marrow-derived precursors. Twenty mice were divided into four groups (n = 5) and treated with DC vaccines, unpulsed mature DCs, Panc02 lysates or no treatment. After tumor induction, mice underwent three magnetic resonance imaging scans to track tumor growth. Apparent diffusion coefficient (ADC), a quantitative magnetic resonance imaging measurement of tumor microstructure, was calculated. Survival was tracked. Tumor tissue was collected after death and stained with hematoxylin and eosin, Masson's trichrome, terminal deoxynucleotidyl transferase dUTP nick end labeling and anti-CD8 stains for histology. RESULTS DC-vaccinated mice demonstrated stronger anti-tumor cytotoxicity compared with control groups on lactate dehydrogenase assay. DC vaccine mice also demonstrated decreased tumor volume, prolonged survival and increased ΔADC compared with control groups. On histology, the DC vaccine group had increased apoptosis, increased CD8+ T cells and decreased collagen. ΔADC negatively correlated with % collagen in tumor tissues. DISCUSSION Prophylactic DC vaccination may inhibit PDAC tumor growth during recurrence and prolong survival. ΔADC may be a potential imaging biomarker that correlates with tumor histological features.
Collapse
Affiliation(s)
- Anna Shangguan
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Medical Student Training Program, Northwestern University, Chicago, Illinois, USA
| | - Na Shang
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Matteo Figini
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Liang Pan
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Radiology, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Jia Yang
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Quanhong Ma
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Su Hu
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Aydin Eresen
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Chong Sun
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Bin Wang
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, China
| | - Yuri Velichko
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Vahid Yaghmai
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Zhuoli Zhang
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA.
| |
Collapse
|
28
|
Fenton SE, Sosman JA, Chandra S. Resistance mechanisms in melanoma to immuneoncologic therapy with checkpoint inhibitors. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:744-761. [PMID: 35582566 PMCID: PMC8992532 DOI: 10.20517/cdr.2019.28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/16/2019] [Accepted: 05/22/2019] [Indexed: 11/14/2022]
Abstract
Checkpoint inhibitors act by blocking physiologic mechanisms coopted by tumor cells to evade immune surveillance, restoring the immune system's ability to identify and kill malignant cells. These therapies have dramatically improved outcomes in multiple tumor types with durable responses in many patients, leading to FDA approval first in advanced melanoma, then in many other malignancies. However, as experience with checkpoint inhibitors has grown, populations of patients who are primary nonresponders or develop secondary resistance have been the majority of cases, even in melanoma. Mechanisms of resistance include those inherent to the tumor microenvironment, the tumor cells themselves, and the function of the patient's native immune cells. This review will discuss resistance to checkpoint inhibitors in melanoma as well as possible methods to restore sensitivity.
Collapse
Affiliation(s)
- Sarah E. Fenton
- Division of Hematology Oncology, Northwestern University, Chicago, IL 60611, USA
| | - Jeffrey A. Sosman
- Division of Hematology Oncology, Northwestern University, Chicago, IL 60611, USA
| | - Sunandana Chandra
- Division of Hematology Oncology, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
29
|
Lin H, Li S, Shu HB. The Membrane-Associated MARCH E3 Ligase Family: Emerging Roles in Immune Regulation. Front Immunol 2019; 10:1751. [PMID: 31404274 PMCID: PMC6669941 DOI: 10.3389/fimmu.2019.01751] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/10/2019] [Indexed: 01/13/2023] Open
Abstract
The membrane-associated RING-CH-type finger (MARCH) proteins of E3 ubiquitin ligases have emerged as critical regulators of immune responses. MARCH proteins target immune receptors, viral proteins as well as components in innate immune response for polyubiquitination and degradations via distinct routes. This review summarizes the current progress about MARCH proteins and their regulation on immune responses.
Collapse
Affiliation(s)
- Heng Lin
- Department of Infectious Diseases, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Shu Li
- Department of Infectious Diseases, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hong-Bing Shu
- Department of Infectious Diseases, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
30
|
Alameddine J, Godefroy E, Papargyris L, Sarrabayrouse G, Tabiasco J, Bridonneau C, Yazdanbakhsh K, Sokol H, Altare F, Jotereau F. Faecalibacterium prausnitzii Skews Human DC to Prime IL10-Producing T Cells Through TLR2/6/JNK Signaling and IL-10, IL-27, CD39, and IDO-1 Induction. Front Immunol 2019; 10:143. [PMID: 30787928 PMCID: PMC6373781 DOI: 10.3389/fimmu.2019.00143] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/17/2019] [Indexed: 12/25/2022] Open
Abstract
The human colonic mucosa contains regulatory type 1-like (Tr1-like, i.e., IL-10-secreting and Foxp3-negative) T cells specific for the gut Clostridium Faecalibacterium prausnitzii (F. prausnitzii), which are both decreased in Crohn's disease patients. These data, together with the demonstration, in mice, that colonic regulatory T cells (Treg) induced by Clostridium bacteria are key players in colon homeostasis, support a similar role for F. prausnitzii-specific Treg in the human colon. Here we assessed the mechanisms whereby F. prausnitzii induces human colonic Treg. We demonstrated that F. prausnitzii, but not related Clostridia, skewed human dendritic cells to prime IL-10-secreting T cells. Accordingly, F. prausnitzii induced dendritic cells to express a unique array of potent Tr1/Treg polarizing molecules: IL-10, IL-27, CD39, IDO-1, and PDL-1 and, following TLR4 stimulation, inhibited their up-regulation of costimulation molecules as well as their production of pro-inflammatory cytokines IL-12 (p35 and p40) and TNFα. We further showed that these potent tolerogenic effects relied on F. prausnitzii-induced TLR2/6 triggering, JNK signaling and CD39 ectonucleotidase activity, which was induced by IDO-1 and IL-27. These data, together with the presence of F. prausnitzii-specific Tr1-like Treg in the human colon, point out to dendritic cells polarization by F. prausnitzii as the first described cellular mechanism whereby the microbiota composition may affect human colon homeostasis. Identification of F. prausnitzii-induced mediators involved in Tr1-like Treg induction by dendritic cells opens therapeutic avenues for the treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Joudy Alameddine
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | | | - Loukas Papargyris
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Angers, France
| | | | - Julie Tabiasco
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Angers, France
| | - Chantal Bridonneau
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | | | - Harry Sokol
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France.,APHP Laboratoire des Biomolécules (LBM), CNRS, INSERM, Sorbonne Universités, Paris, France.,Laboratoire des Biomolécules, Département de Chimie, CNRS, PSL Research University, Paris, France
| | - Frédéric Altare
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Francine Jotereau
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| |
Collapse
|
31
|
Kerperien J, Veening-Griffioen D, Wehkamp T, van Esch BCAM, Hofman GA, Cornelissen P, Boon L, Jeurink PV, Garssen J, Knippels LMJ, Willemsen LEM. IL-10 Receptor or TGF-β Neutralization Abrogates the Protective Effect of a Specific Nondigestible Oligosaccharide Mixture in Cow-Milk-Allergic Mice. J Nutr 2018; 148:1372-1379. [PMID: 29986071 PMCID: PMC6074790 DOI: 10.1093/jn/nxy104] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/19/2018] [Accepted: 04/20/2018] [Indexed: 11/14/2022] Open
Abstract
Background Dietary nondigestible, short-chain galacto-, long-chain fructo-, and pectin-derived acidic oligosaccharides (GFAs) lower the effector response in cow-milk-allergic (CMA) mice; and forkhead box P3 (Foxp3)-positive regulatory T cells (Tregs) were shown to contribute to this. Objective The aim of this study was to assess the contribution of interleukin 10 (IL-10) and transforming growth factor β (TGF-β) to the protective effect of the GFA diet in CMA mice. Methods Female C3H/HeOuJ mice, 3-4 wk old, were orally sensitized with cholera toxin (Sham) or whey and cholera toxin (Whey) 1 time/wk for 5 consecutive weeks and challenged with whey 1 wk later. The mice were fed a control or 1% GFA (9:2:1) (Whey+GFA) diet starting 2 wk before the first sensitization. In a second experiment, the mice were also injected with αIL-10 receptor (αIL-10r), αTGF-β, or isotype control antibodies 24 h before each sensitization. The acute allergic skin response, anaphylaxis score, whey-specific IgE, mucosal mast cell protease 1 (mMCP-1), and Treg frequency in the mesenteric lymph nodes (MLNs) and intestinal Foxp3, Il10, and Tgfb mRNA expression were determined. Results In Whey+GFA mice, intestinal Il10, Tgfb, or Foxp3 mRNA expression was 2-10 times higher (P < 0.05) and the MLN Treg frequency was 25% higher compared with Whey mice (P < 0.05). The acute allergic skin response was 50% lower in Whey+GFA mice compared with Whey mice (P < 0.01), and IL-10 receptor (IL-10r) or TGF-β neutralizing antibodies prevented this protective effect (P < 0.001). The Whey mice had higher serum mMCP-1 concentrations and whey-immunoglobulin E (-IgE) levels than Sham mice (P < 0.01), whereas these were not higher in Whey+GFA mice, and neutralizing antibodies partially interfered with these responses. Conclusions Dietary GFAs enhance the Treg frequency in the MLNs and mucosal IL-10 and TGF-β transcription while suppressing the allergic effector response. Neutralizing antibodies showed that the allergy-protective effect of the GFA diet was mediated by IL-10 and TGF-β in CMA mice.
Collapse
Affiliation(s)
- JoAnn Kerperien
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Désirée Veening-Griffioen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | | | - Betty C A M van Esch
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Gerard A Hofman
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Paquita Cornelissen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | | | - Prescilla V Jeurink
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Leon M J Knippels
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Linette E M Willemsen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
32
|
March1 E3 Ubiquitin Ligase Modulates Features of Allergic Asthma in an Ovalbumin-Induced Mouse Model of Lung Inflammation. J Immunol Res 2018; 2018:3823910. [PMID: 29854835 PMCID: PMC5960577 DOI: 10.1155/2018/3823910] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/08/2018] [Indexed: 01/13/2023] Open
Abstract
Membrane-associated RING-CH-1 (March1) is a member of the March family of E3 ubiquitin ligases. March1 downregulates cell surface expression of MHC II and CD86 by targeting them to lysosomal degradation. Given the key roles of MHC class II and CD86 in T cell activation and to get further insights into the development of allergic inflammation, we asked whether March1 deficiency exacerbates or attenuates features of allergic asthma in mice. Herein, we used an acute model of allergy to compare the asthmatic phenotype of March1-deficient and -sufficient mice immunized with ovalbumin (OVA) and later challenged by intranasal instillation of OVA in the lungs. We found that eosinophilic inflammation in airways and lung tissue was similar between WT and March1-/- allergic mice, whereas neutrophilic inflammation was significant only in March1-/- mice. Airway hyperresponsiveness as well as levels of IFN-γ, IL-13, IL-6, and IL-10 was lower in the lungs of asthmatic March1-/- mice compared to WT, whereas lung levels of TNF-α, IL-4, and IL-5 were not significantly different. Interestingly, in the serum, levels of total and ova-specific IgE were reduced in March1-deficient mice as compared to WT mice. Taken together, our results demonstrate a role of March1 E3 ubiquitin ligase in modulating allergic responses.
Collapse
|
33
|
Stadler S, Singh VK, Knörr F, Damm-Welk C, Woessmann W. Immune Response against ALK in Children with ALK-Positive Anaplastic Large Cell Lymphoma. Cancers (Basel) 2018; 10:cancers10040114. [PMID: 29642597 PMCID: PMC5923369 DOI: 10.3390/cancers10040114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/05/2018] [Accepted: 04/07/2018] [Indexed: 12/26/2022] Open
Abstract
Patients with anaplastic lymphoma kinase (ALK)-positive anaplastic large cell lymphoma (ALCL) mount a humoral and cellular immune response against ALK. More than 90% of children and adolescents with ALK-positive ALCL have detectable anti-ALK antibodies in serum or plasma, and the antibody titer inversely correlates with the risk of relapse. ALK-specific CD8 and CD4 T cell responses have been described in patients with ALK-positive ALCL. Vaccination with ALK DNA led to protection against lymphoma growth in a murine model. Collectively, these data suggest that the ALK-specific immune response is involved in the control of the disease. The characteristics of the humoral and cellular immune response against ALK as well as tumor immune escape mechanisms have been increasingly investigated. However, tumor and host factors contributing to the individual immune response against ALK are still largely unknown. Depending on the individual strength of the immune response and its determinants, individualized immunological approaches might be appropriate for the consolidation of ALCL patients. Strategies such as ALK vaccination could be effective for those with a pre-existing anti-tumor immunity, while an allogeneic blood stem cell transplantation or check-point inhibition could be effective for others.
Collapse
Affiliation(s)
- Serena Stadler
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, D-35392 Giessen, Germany.
| | - Vijay Kumar Singh
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, D-35392 Giessen, Germany.
| | - Fabian Knörr
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, D-35392 Giessen, Germany.
| | - Christine Damm-Welk
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, D-35392 Giessen, Germany.
| | - Wilhelm Woessmann
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, D-35392 Giessen, Germany.
| |
Collapse
|
34
|
Bashaw AA, Leggatt GR, Chandra J, Tuong ZK, Frazer IH. Modulation of antigen presenting cell functions during chronic HPV infection. PAPILLOMAVIRUS RESEARCH (AMSTERDAM, NETHERLANDS) 2017; 4:58-65. [PMID: 29179871 PMCID: PMC5883240 DOI: 10.1016/j.pvr.2017.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/14/2017] [Accepted: 08/15/2017] [Indexed: 12/12/2022]
Abstract
High-risk human papillomaviruses (HR-HPV) infect basal keratinocytes, where in some individuals they evade host immune responses and persist. Persistent HR-HPV infection of the cervix causes precancerous neoplasia that can eventuate in cervical cancer. Dendritic cells (DCs) are efficient in priming/cross-priming antigen-specific T cells and generating antiviral and antitumor cytotoxic CD8+ T cells. However, HR-HPV have adopted various immunosuppressive strategies, with modulation of DC function crucial to escape from the host adaptive immune response. HPV E6 and E7 oncoproteins alter recruitment and localization of epidermal DCs, while soluble regulatory factors derived from HPV-induced hyperplastic epithelium change DC development and influence initiation of specific cellular immune responses. This review focuses on current evidence for HR-HPV manipulation of antigen presentation in dendritic cells and escape from host immunity.
Collapse
Affiliation(s)
- Abate Assefa Bashaw
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Graham R Leggatt
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Janin Chandra
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Zewen K Tuong
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Ian H Frazer
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia.
| |
Collapse
|
35
|
Poujol F, Monneret G, Gallet-Gorius E, Pachot A, Textoris J, Venet F. Ex vivo Stimulation of Lymphocytes with IL-10 Mimics Sepsis-Induced Intrinsic T-Cell Alterations. Immunol Invest 2017; 47:154-168. [PMID: 29182416 DOI: 10.1080/08820139.2017.1407786] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Profound T-cell alterations are observed in septic patients in association with increased risk of secondary infection and mortality. The pathophysiological mechanisms leading to such dysfunctions are not completely understood and direct and indirect mechanisms have been described. In this study we evaluated whether ex vivo stimulation of lymphocytes with IL-10, an immunosuppressive cytokine released at the systemic level during sepsis, could mimic sepsis-induced intrinsic T-cell alterations. We showed that recombinant human IL-10 priming of T cells altered their proliferative response to anti-CD2/CD3/CD28 antibody-coated beads and PHA stimulations, in a dose-dependent manner independently of accessory cells. This priming also significantly decreased T-cell secretion of IL-2 and IFNγ following stimulation. Furthermore, we demonstrated that IL-10 reduction of T-cell functionality was associated with increased FOXP3 expression in CD4+CD25+CD127- regulatory T cells as observed in sepsis. Finally, we found that blocking the increased IL-10 concentration in plasma from septic shock patients increased the proliferative response of responding T cells from healthy controls. We describe here an ex vivo model recapitulating features of sepsis-induced intrinsic T-cell alterations. This should help, in further studies, to decipher the pathophysiological mechanisms of T-cell alterations induced after septic shock.
Collapse
Affiliation(s)
- Fanny Poujol
- a Joint Research Unit, EA 7426 bioMérieux/Hospices Civils de Lyon/UCBL1 "Pathophysiology of injury induced immunosuppression (PI3)", Edouard Herriot Hospital , Lyon , France.,b Immunology Laboratory, Joint Research Unit (BioMérieux/Hospices Civils de Lyon/UCBL1), Edouard Herriot Hospital , Lyon , France
| | - Guillaume Monneret
- a Joint Research Unit, EA 7426 bioMérieux/Hospices Civils de Lyon/UCBL1 "Pathophysiology of injury induced immunosuppression (PI3)", Edouard Herriot Hospital , Lyon , France.,c Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Immunology Laboratory, Edouard Herriot Hospital , Lyon , France
| | - Emmanuelle Gallet-Gorius
- a Joint Research Unit, EA 7426 bioMérieux/Hospices Civils de Lyon/UCBL1 "Pathophysiology of injury induced immunosuppression (PI3)", Edouard Herriot Hospital , Lyon , France.,b Immunology Laboratory, Joint Research Unit (BioMérieux/Hospices Civils de Lyon/UCBL1), Edouard Herriot Hospital , Lyon , France
| | - Alexandre Pachot
- a Joint Research Unit, EA 7426 bioMérieux/Hospices Civils de Lyon/UCBL1 "Pathophysiology of injury induced immunosuppression (PI3)", Edouard Herriot Hospital , Lyon , France.,b Immunology Laboratory, Joint Research Unit (BioMérieux/Hospices Civils de Lyon/UCBL1), Edouard Herriot Hospital , Lyon , France
| | - Julien Textoris
- a Joint Research Unit, EA 7426 bioMérieux/Hospices Civils de Lyon/UCBL1 "Pathophysiology of injury induced immunosuppression (PI3)", Edouard Herriot Hospital , Lyon , France.,b Immunology Laboratory, Joint Research Unit (BioMérieux/Hospices Civils de Lyon/UCBL1), Edouard Herriot Hospital , Lyon , France.,d Anesthesia and Critical Care Medicine Department , Hospices Civils de Lyon, Burn ICU, Edouard Herriot Hospital , Lyon , France
| | - Fabienne Venet
- a Joint Research Unit, EA 7426 bioMérieux/Hospices Civils de Lyon/UCBL1 "Pathophysiology of injury induced immunosuppression (PI3)", Edouard Herriot Hospital , Lyon , France.,c Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Immunology Laboratory, Edouard Herriot Hospital , Lyon , France
| |
Collapse
|
36
|
Gide TN, Wilmott JS, Scolyer RA, Long GV. Primary and Acquired Resistance to Immune Checkpoint Inhibitors in Metastatic Melanoma. Clin Cancer Res 2017; 24:1260-1270. [PMID: 29127120 DOI: 10.1158/1078-0432.ccr-17-2267] [Citation(s) in RCA: 303] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 10/15/2017] [Accepted: 11/06/2017] [Indexed: 11/16/2022]
Abstract
Immune checkpoint inhibitors have revolutionized the treatment of patients with advanced-stage metastatic melanoma, as well as patients with many other solid cancers, yielding long-lasting responses and improved survival. However, a subset of patients who initially respond to immunotherapy, later relapse and develop therapy resistance (termed "acquired resistance"), whereas others do not respond at all (termed "primary resistance"). Primary and acquired resistance are key clinical barriers to further improving outcomes of patients with metastatic melanoma, and the known mechanisms underlying each involves various components of the cancer immune cycle, and interactions between multiple signaling molecules and pathways. Due to this complexity, current knowledge on resistance mechanisms is still incomplete. Overcoming therapy resistance requires a thorough understanding of the mechanisms underlying immune evasion by tumors. In this review, we explore the mechanisms of primary and acquired resistance to immunotherapy in melanoma and detail potential therapeutic strategies to prevent and overcome them. Clin Cancer Res; 24(6); 1260-70. ©2017 AACR.
Collapse
Affiliation(s)
- Tuba N Gide
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Royal North Shore Hospital, Sydney, NSW, Australia
- Mater Hospital, North Sydney, NSW, Australia
| |
Collapse
|
37
|
Schildberg FA, Klein SR, Freeman GJ, Sharpe AH. Coinhibitory Pathways in the B7-CD28 Ligand-Receptor Family. Immunity 2017; 44:955-72. [PMID: 27192563 DOI: 10.1016/j.immuni.2016.05.002] [Citation(s) in RCA: 431] [Impact Index Per Article: 53.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Indexed: 01/10/2023]
Abstract
Immune responses need to be controlled for optimal protective immunity and tolerance. Coinhibitory pathways in the B7-CD28 family provide critical inhibitory signals that regulate immune homeostasis and defense and protect tissue integrity. These coinhibitory signals limit the strength and duration of immune responses, thereby curbing immune-mediated tissue damage, regulating resolution of inflammation, and maintaining tolerance to prevent autoimmunity. Tumors and microbes that cause chronic infections can exploit these coinhibitory pathways to establish an immunosuppressive microenvironment, hindering their eradication. Advances in understanding T cell coinhibitory pathways have stimulated a new era of immunotherapy with effective drugs to treat cancer, autoimmune and infectious diseases, and transplant rejection. In this review we discuss the current knowledge of the mechanisms underlying the coinhibitory functions of pathways in the B7-CD28 family, the diverse functional consequences of these inhibitory signals on immune responses, and the overlapping and unique functions of these key immunoregulatory pathways.
Collapse
Affiliation(s)
- Frank A Schildberg
- Department of Microbiology and Immunobiology, and Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah R Klein
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology, and Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
38
|
Li L, Luo Z, Song Z, Zheng L, Chen T. Pre-transplant infusion of donor-derived dendritic cells maintained at the immature stage by sinomenine increases splenic Foxp3 + Tregs in recipient rats after renal allotransplantation. Transpl Immunol 2017; 45:22-28. [PMID: 28802587 DOI: 10.1016/j.trim.2017.08.004] [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: 04/07/2017] [Revised: 08/07/2017] [Accepted: 08/08/2017] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The immunosuppressive mechanism of sinomenine in organ allotransplantation was investigated, especially its effect of blocking dendritic cell (DC) maturation, which might influence the frequency of regulatory T cells (Tregs). METHODS Bone marrow cells from male donor Wistar rats were induced to differentiate into DCs in vitro in the presence or absence of sinomenine, and characterized by flow cytometry. These two groups of DCs were respectively injected into male recipient Sprague-Dawley rats via the tail vein, at both high and low doses. Sprague-Dawley rats receiving saline injection were used as controls. Seven days later, renal transplantation was performed from donor Wistar rats to the recipient Sprague-Dawley rats. Seven days after transplantation, spleens were collected from the recipients. The proportions of Tregs and Foxp3+ Tregs to CD4+ T cells were determined using flow cytometry. RESULTS With sinomenine treatment, the frequency of mature DCs was reduced, as indicated by lower expression of the surface markers CD80, CD86, and RT1B. In recipient Sprague-Dawley rats that received sinomenine-treated DCs before renal allotransplantation, the proportions of splenic Tregs and Foxp3+ Tregs were significantly higher than in control recipients receiving saline or DCs without sinomenine treatment (all p<0.05). A high dose of sinomenine-treated DCs (106 cells) had a more obvious effect in increasing Tregs than the low dose (105 cells) (p<0.05). CONCLUSION Pre-transplant infusion of donor-derived sinomenine-induced maturation arrested DCs could result in the increase of Foxp3+ Tregs in the spleens of recipients after renal allotransplantation.
Collapse
Affiliation(s)
- Lian Li
- Department of Urology, The Second Hospital, University of South China, Hengyang, Hunan, China
| | - Zhigang Luo
- Department of Urology, The Second Hospital, University of South China, Hengyang, Hunan, China.
| | - Zhe Song
- Department of Urology, The Second Hospital, University of South China, Hengyang, Hunan, China
| | - Liwen Zheng
- Department of Plastic Surgery, Hengyang No.1 People's Hospital, Hengyang, Hunan, China
| | - Tuo Chen
- Department of Urology, The Second People's Hospital of Yueyang, Yueyang, Hunan, China
| |
Collapse
|
39
|
Dendritic cells conditioned by activin A-induced regulatory T cells exhibit enhanced tolerogenic properties and protect against experimental asthma. J Allergy Clin Immunol 2017; 141:671-684.e7. [PMID: 28579377 DOI: 10.1016/j.jaci.2017.03.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 02/14/2017] [Accepted: 03/23/2017] [Indexed: 11/21/2022]
Abstract
BACKGROUND Previously, we demonstrated that regulatory T (Treg) cells induced by the cytokine activin-A suppress TH2-mediated allergic responses and linked airway disease. Still, the effects of activin-A-induced regulatory T (Act-A-iTreg) cells on the regulation of dendritic cell (DC)-driven allergic inflammation remain elusive. OBJECTIVE Here we investigated whether Act-A-iTreg cells can modulate DC responses and endow them with enhanced tolerogenic functions. METHODS Using adoptive cell transfer studies in mouse models of allergic airway disease, we examined the effects of Act-A-iTreg cells on DC phenotype, maturation status, and TH2 cell priming potential. Genome-wide gene expression profiling characterized the transcriptional networks induced in tolerogenic DCs by Act-A-iTreg cells. The ability of DCs conditioned by Act-A-iTreg cells (Act-A-iTreg cell-modified DCs) to protect against experimental asthma, and the mechanisms involved were also explored. RESULTS Act-A-iTreg cell-modified DCs exhibited a significantly impaired capacity to uptake allergen and stimulate naive and TH2 effector responses on allergen stimulation in vivo accompanied by markedly attenuated inflammatory cytokine release in response to LPS. Gene-profiling studies revealed that Act-A-iTreg cells dampened crucial TH2-skewing transcriptional networks in DCs. Administration of Act-A-iTreg cell-modified DCs ameliorated cardinal asthma manifestations in preventive and therapeutic protocols through generation of strongly suppressive forkhead box P3+ Treg cells. Finally, programed death protein 1/programmed death ligand 1 signaling pathways were essential in potentiating the generation of DCs with tolerogenic properties by Act-A-iTreg cells. CONCLUSION Our studies reveal that Act-A-iTreg cells instruct the generation of a highly effective immunoregulatory circuit encompassing tolerogenic DCs and forkhead box P3+ Treg cells that could be targeted for the design of novel immunotherapies for allergic disorders.
Collapse
|
40
|
De Angelis Rigotti F, De Gassart A, Pforr C, Cano F, N'Guessan P, Combes A, Camossetto V, Lehner PJ, Pierre P, Gatti E. MARCH9-mediated ubiquitination regulates MHC I export from the TGN. Immunol Cell Biol 2017; 95:753-764. [PMID: 28559542 DOI: 10.1038/icb.2017.44] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 01/08/2023]
Abstract
Given the heterogeneous nature of antigens, major histocompatibility complex class I (MHC I) intracellular transport intersects with multiple degradation pathways for efficient peptide loading and presentation to cytotoxic T cells. MHC I loading with peptides in the endoplasmic reticulum (ER) is a tightly regulated process, while post-ER intracellular transport is considered to occur by default, leading to peptide-bearing MHC I delivery to the plasma membrane. We show here that MHC I traffic is submitted to a previously uncharacterized sorting step at the trans Golgi network (TGN), dependent on the ubiquitination of its cytoplasmic tail lysine residues. MHC I ubiquitination is mediated by the E3 ligase membrane-associated RING-CH 9 (MARCH9) and allows MHC I access to Syntaxin 6-positive endosomal compartments. We further show that MARCH9 can also target the human MHC I-like lipid antigen-presentation molecule CD1a. MARCH9 expression is modulated by microbial pattern exposure in dendritic cells (DCs), thus revealing the role of this ubiquitin E3 ligase in coordinating MHC I access to endosomes and DC activation for efficient antigen cross-presentation.
Collapse
Affiliation(s)
| | - Aude De Gassart
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, CNRS, INSERM, Marseille, France
| | - Carina Pforr
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, CNRS, INSERM, Marseille, France
| | - Florencia Cano
- Division of Immunology, Cambridge Institute of Medical Research, Addenbrookes Hospital, Cambridge, UK
| | - Prudence N'Guessan
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, CNRS, INSERM, Marseille, France
| | - Alexis Combes
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, CNRS, INSERM, Marseille, France
| | - Voahirana Camossetto
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, CNRS, INSERM, Marseille, France
| | - Paul J Lehner
- Division of Immunology, Cambridge Institute of Medical Research, Addenbrookes Hospital, Cambridge, UK
| | - Philippe Pierre
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, CNRS, INSERM, Marseille, France
| | - Evelina Gatti
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, CNRS, INSERM, Marseille, France
| |
Collapse
|
41
|
Seng A, Yankee TM. The Role of the Ikaros Family of Transcription Factors in Regulatory T cell Development and Function. ACTA ACUST UNITED AC 2017; 8. [PMID: 28758047 PMCID: PMC5531196 DOI: 10.4172/2155-9899.1000495] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Regulatory T (Treg) cells are a subset of immune cells that maintain homeostasis by promoting immune tolerance and suppressing the immune response via a variety of mechanisms such as secreting cytokines, killing reactive immune cells, and inducing anergy. Dysfunction of Treg cells has been implicated in inflammatory diseases such as autoimmunity and transplant rejection. Conversely, too many or hyperresponsive Treg cells has been observed in cancer and chronic infections. Treg cells have proven to be difficult to study as there are no definitive Treg surface markers. Additionally, Tregs can gain pro-inflammatory phenotype depending on stimuli. In this commentary, we discuss the expression and function of members of the Ikaros family of transcription factors during Treg cell development and activation.
Collapse
Affiliation(s)
- Amara Seng
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Thomas M Yankee
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
42
|
Wallberg M, Recino A, Phillips J, Howie D, Vienne M, Paluch C, Azuma M, Wong FS, Waldmann H, Cooke A. Anti-CD3 treatment up-regulates programmed cell death protein-1 expression on activated effector T cells and severely impairs their inflammatory capacity. Immunology 2017; 151:248-260. [PMID: 28211040 PMCID: PMC5418468 DOI: 10.1111/imm.12729] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/03/2017] [Accepted: 02/13/2017] [Indexed: 01/07/2023] Open
Abstract
T cells play a key role in the pathogenesis of type 1 diabetes, and targeting the CD3 component of the T‐cell receptor complex provides one therapeutic approach. Anti‐CD3 treatment can reverse overt disease in spontaneously diabetic non‐obese diabetic mice, an effect proposed to, at least in part, be caused by a selective depletion of pathogenic cells. We have used a transfer model to further investigate the effects of anti‐CD3 treatment on green fluorescent protein (GFP)+ islet‐specific effector T cells in vivo. The GFP expression allowed us to isolate the known effectors at different time‐points during treatment to assess cell presence in various organs as well as gene expression and cytokine production. We find, in this model, that anti‐CD3 treatment does not preferentially deplete the transferred effector cells, but instead inhibits their metabolic function and their production of interferon‐γ. Programmed cell death protein 1 (PD‐1) expression was up‐regulated on the effector cells from anti‐CD3‐treated mice, and diabetes induced through anti‐PD‐L1 antibody could only be reversed with anti‐CD3 antibody if the anti‐CD3 treatment lasted beyond the point when the anti‐PD‐L1 antibody was washed out of the system. This suggests that PD‐1/PD‐L1 interaction plays an important role in the anti‐CD3 antibody mediated protection. Our data demonstrate an additional mechanism by which anti‐CD3 therapy can reverse diabetogenesis.
Collapse
Affiliation(s)
- Maja Wallberg
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Asha Recino
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Jenny Phillips
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Duncan Howie
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Margaux Vienne
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | - Miyuki Azuma
- Department of Molecular Immunology Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - F Susan Wong
- Diabetes Research Group, Institute of Molecular and Experimental Medicine, Cardiff School of Medicine, Cardiff University, Cardiff, UK
| | - Herman Waldmann
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Anne Cooke
- Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
43
|
Akkaya B, Holstein AH, Isaac C, Maz MP, Glass DD, Shevach EM, Akkaya M. Ex-vivo iTreg differentiation revisited: Convenient alternatives to existing strategies. J Immunol Methods 2016; 441:67-71. [PMID: 27919837 DOI: 10.1016/j.jim.2016.11.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 11/26/2016] [Accepted: 11/30/2016] [Indexed: 11/26/2022]
Abstract
Ex-vivo differentiation of regulatory T cells (Tregs) from naïve CD4+ T-cells has been widely used in immunological research. Isolation of a highly pure naïve T cell population is the key factor that determines the efficiency of subsequent Treg differentiation. Currently, this step relies mostly on FACS sorting, which is often costly, time consuming, and inconvenient. Alternatively, magnetic separation of T-cells can be performed; yet, available protocols fail to reach sort level purity and consequently result in low Treg differentiation efficiency. Here, we present the results of a comprehensive side-by-side comparison of various magnetic separation strategies and FACS sorting in multiple levels. Additionally, we propose a novel optimized custom made magnetic separation protocol, which not only yields sort level purity and Treg differentiation but also lowers the reagent costs up to 75% compared to the commercially available purification kits. The highly pure naïve CD4+ T-cell population obtained by this versatile method can also be used for differentiation of other T-cell subsets; therefore this protocol may have broad applications in T-cell research.
Collapse
Affiliation(s)
- Billur Akkaya
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Amanda H Holstein
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Christopher Isaac
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Mitra P Maz
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Deborah D Glass
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Ethan M Shevach
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Munir Akkaya
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, United States.
| |
Collapse
|
44
|
Mavin E, Nicholson L, Rafez Ahmed S, Gao F, Dickinson A, Wang XN. Human Regulatory T Cells Mediate Transcriptional Modulation of Dendritic Cell Function. THE JOURNAL OF IMMUNOLOGY 2016; 198:138-146. [PMID: 27895173 DOI: 10.4049/jimmunol.1502487] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 10/21/2016] [Indexed: 12/21/2022]
Abstract
Regulatory T cells (Treg) attenuate dendritic cell (DC) maturation and stimulatory function. Current knowledge on the functional impact of semimature DC is limited to CD4+ T cell proliferation and cytokine production. Little is known about the molecular basis underpinning the functional effects of Treg-treated DC (Treg-DC). We present novel evidence that Treg-DC skewed CD4+ naive T cell polarization toward a regulatory phenotype and impaired CD8+ T cell allo-reactive responses, including their ability to induce target tissue damage in a unique in vitro human graft-versus-host disease skin explant model. Microarray analysis clustered Treg-DC as a discrete population from mature-DC and immature-DC, with 51 and 93 genes that were significantly over- or underexpressed, respectively, compared with mature-DC. Quantitative real-time PCR analysis revealed an intermediate expression level of CD38, CD83, CD80 and CD86 mRNA in Treg-DC, lower than mature-DC, higher than immature-DC. We also observed an attenuation of NF-κB pathway, an upstream regulator of the aforementioned genes, concomitant with reduced expression of two NF-κB-signaling related genes RELB and NFκBIZ, in the Treg-DC, together with an increased expression of Wnt5a, a negative regulator of DC differentiation. We further confirmed that the Treg-DC-mediated skewed CD4+ naive T cell polarization resulted from decreased IL-12 secretion by Treg-DC, which may be post-transcriptionally modulated by decreased expression of microRNA-155 in Treg-DC. To our knowledge, this is the first study demonstrating a transcriptional modulation of DC function by human Treg, partially via attenuation of the NF-κB signaling pathway and upregulation of Wnt5a, suggesting Treg may interfere with DC reprogramming during maturation, thereby modulating DC function.
Collapse
Affiliation(s)
- Emily Mavin
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom; and
| | - Lindsay Nicholson
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom; and
| | - Syed Rafez Ahmed
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom; and
| | - Fei Gao
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE7 1RU, United Kingdom
| | - Anne Dickinson
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom; and
| | - Xiao-Nong Wang
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom; and
| |
Collapse
|
45
|
Meng Y, Hu J, Chen Y, Yu T, Hu L. Silencing MARCH1 suppresses proliferation, migration and invasion of ovarian cancer SKOV3 cells via downregulation of NF-κB and Wnt/β-catenin pathways. Oncol Rep 2016; 36:2463-2470. [PMID: 27633480 PMCID: PMC5055210 DOI: 10.3892/or.2016.5076] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/27/2016] [Indexed: 11/25/2022] Open
Abstract
Membrane-associated RING-CH (MARCH) belongs to the family of RING-CH type E3 ubiquitin ligases. MARCH1 ubiquitinates and downregulates MHC class II expression in APCs and targets major players of the immune system. However, the role of MARCH1 in ovarian cancer has not been elucidated. The present study investigated the function of MARCH1 in ovarian cancer and the potential mechanisms involved. MARCH1 expression was examined in human ovarian cancer tissue specimens by immunohistochemistry. The role of MARCH1 in ovarian cancer cells was assessed by cell proliferation, migration and invasion assays with MARCH1 gene silencing. To investigate the mechanism by which MARCH1 functions, correlation between MARCH1 and the cell signaling pathways were analyzed using a luciferase reporter assay, real-time RT-PCR, western blot assay and immunofluorescence. MARCH1 was found to be overexpressed in ovarian cancer tissues when compared to adjacent non-tumor and normal ovarian tissues. Silencing MARCH1 inhibited SKOV3 cell proliferation, invasion and migration, as well as inhibiting the NF-κB and the Wnt/β-catenin pathways. MARCH1 functions as a tumor promoter by upregulating the NF-κB and the Wnt/β-catenin pathways, indicating that MARCH1 may be a therapeutic target for patients with ovarian cancer.
Collapse
Affiliation(s)
- Ying Meng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jianguo Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yuhong Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Tinghe Yu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Lina Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
46
|
Santegoets SJAM, Welters MJP, van der Burg SH. Monitoring of the Immune Dysfunction in Cancer Patients. Vaccines (Basel) 2016; 4:vaccines4030029. [PMID: 27598210 PMCID: PMC5041023 DOI: 10.3390/vaccines4030029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/25/2016] [Accepted: 08/27/2016] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy shows promising clinical results in patients with different types of cancer, but its full potential is not reached due to immune dysfunction as a result of several suppressive mechanisms that play a role in cancer development and progression. Monitoring of immune dysfunction is a prerequisite for the development of strategies aiming to alleviate cancer-induced immune suppression. At this point, the level at which immune dysfunction occurs has to be established, the underlying mechanism(s) need to be known, as well as the techniques to assess this. While it is relatively easy to measure general signs of immune suppression, it turns out that accurate monitoring of the frequency and function of immune-suppressive cells is still difficult. A lack of truly specific markers, the phenotypic complexity among suppressive cells of the same lineage, but potentially with different functions and functional assays that may not cover every mechanistic aspect of immune suppression are among the reasons complicating proper assessments. Technical innovations in flow and mass cytometry will allow for more complete sets of markers to precisely determine phenotype and associated function. There is, however, a clear need for functional assays that recapitulate more of the mechanisms employed to suppress the immune system.
Collapse
Affiliation(s)
- Saskia J A M Santegoets
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Marij J P Welters
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
47
|
Oh J, Shin JS. Molecular mechanism and cellular function of MHCII ubiquitination. Immunol Rev 2016; 266:134-44. [PMID: 26085212 DOI: 10.1111/imr.12303] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The major histocompatibility complex class II (MHCII) is ubiquitinated via the evolutionarily conserved lysine in the cytoplasmic tail of the β chain in dendritic cells (DCs) and B cells. The ubiquitination is mediated by the membrane-associated RING-CH1 (MARCH1) ubiquitin ligase although it can be also mediated by the homologous ligase MARCH8 in model cell lines. The ubiquitination promotes MHCII endocytosis and lysosomal sorting that results in a reduction in the level of MHCII at cell surface. Functionally, MHCII ubiquitination serves as a means by which DCs suppress MHCII expression and reduce antigen presentation in response to the immune regulatory cytokine interleukin-10 (IL-10) and regulatory T cells. Recently, additional roles of MHCII ubiquitination have emerged. MHCII ubiquitination promoted DC production of inflammatory cytokines in response to the Toll-like receptor ligands. It also potentiated DC ability to activate antigen-specific naive CD4(+) T cells while limiting the amount of antigens presented at cell surface. Similarly, MHCII ubiquitination promoted DC activation of CD4(+) thymocytes supporting regulatory T-cell development independent of its effect of limiting antigen presentation. Thus, ubiquitination appears to confer MHCII a function independent of presenting antigens by a mechanism yet to be identified.
Collapse
Affiliation(s)
- Jaehak Oh
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Jeoung-Sook Shin
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
48
|
Wang JZ, Zhang YH, Guo XH, Zhang HY, Zhang Y. The double-edge role of B cells in mediating antitumor T-cell immunity: Pharmacological strategies for cancer immunotherapy. Int Immunopharmacol 2016; 36:73-85. [PMID: 27111515 DOI: 10.1016/j.intimp.2016.04.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 04/10/2016] [Accepted: 04/13/2016] [Indexed: 12/22/2022]
Abstract
Emerging evidence reveals the controversial role of B cells in antitumor immunity, but the underlying mechanisms have to be explored. Three latest articles published in the issue 521 of Nature in 2015 reconfirmed the puzzling topic and put forward some explanations of how B cells regulate antitumor T-cell responses both positively and negatively. This paper attempts to demonstrate that different B-cell subpopulations have distinct immunological properties and that they are involved in either antitumor responses or immunosuppression. Recent studies supporting the positive and negative roles of B cells in tumor development were summarized comprehensively. Several specific B-cell subpopulations, such as IgG(+), IgA(+), IL-10(+), and regulatory B cells, were described in detail. The mechanisms underlying the controversial B-cell effects were mainly attributed to different B-cell subpopulations, different B-cell-derived cytokines, direct B cell-T cell interaction, different cancer categories, and different malignant stages, and the immunological interaction between B cells and T cells is mediated by dendritic cells. Promising B-cell-based antitumor strategies were proposed and novel B-cell regulators were summarized to present interesting therapeutic targets. Future investigations are needed to make sure that B-cell-based pharmacological strategies benefit cancer immunotherapy substantially.
Collapse
Affiliation(s)
- Jing-Zhang Wang
- Department of Medical Technology, College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China.
| | - Yu-Hua Zhang
- Department of Library, Hebei University of Engineering, Handan 056038, PR China
| | - Xin-Hua Guo
- Department of Medicine, College of Medicine, Hebei University of Engineering, Handan 056002, PR China
| | - Hong-Yan Zhang
- Department of Medical Technology, College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China
| | - Yuan Zhang
- Department of Medical Technology, College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China
| |
Collapse
|
49
|
Zhou J, Ma P, Li J, Cui X, Song W. Improvement of the cytotoxic T lymphocyte response against hepatocellular carcinoma by transduction of cancer cells with an adeno-associated virus carrying the interferon-γ gene. Mol Med Rep 2016; 13:3197-205. [PMID: 26936017 DOI: 10.3892/mmr.2016.4884] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 12/11/2015] [Indexed: 11/06/2022] Open
Abstract
Dendritic cell (DC)-based antigen-targeted immunotherapy may offer effective adjuvant therapy for hepatocellular carcinoma (HCC), in which cytotoxic T lymphocytes (CTLs) are key. However, in a number of cases, the activity of CTLs is completely inhibited due to the downregulated expression of major human leukocyte antigen (HLA) class I molecules by HCC cells. The aim of the present study was to overcome this issue. Hep3B cells were transduced by HCC‑specific recombinant adeno‑associated virus (rAAV) carrying human α‑fetoprotein promoter (AFPp) and the interferon‑γ (IFN‑γ) gene (rAAV/AFPp‑IFN‑γ). rAAV carrying the cytomegalovirus promoter (CMVp) and human α‑fetoprotein (AFP) gene (rAAV/CMVp‑AFP) was used to transduce professional antigen‑presenting DCs for the purpose of stimulating a CTL response. It was observed that transduction of DCs with rAAV/CMVp‑AFP resulted in: (i) AFP and interleukin‑12 expression; (ii) high expression levels of cluster of differentiation (CD)80, CD83, CD86, CD40, HLA‑death receptor and CD1a; (iii) T cell populations with marked IFN‑γ expression; (iv) a high percentage of CD69+/CD8+ T cells; and (v) the activity of CTLs against HLA‑A2‑expressing Hep3B cells. The transduction of Hep3B cells with rAAV/AFPp‑IFN‑γ resulted in: (i) IFN‑γ expression; (ii) upregulated expression of HLA‑A2; and (iii) an improved CTL response against HLA‑A2‑deficient Hep3B cells. rAAV/CMVp‑AFP‑transduced DCs elicited an AFP‑specific and HLA‑class I‑restricted CTL response against Hep3B cells. In conclusion, it was shown that the transduction of Hep3B with rAAV/AFPp-IFN-γ upregulated the expression of HLA-A2 and improved the sensitivity to CTL response.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Ping Ma
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Jun Li
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xiaonan Cui
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Wei Song
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
50
|
Pioli PD, Whiteside SK, Weis JJ, Weis JH. Snai2 and Snai3 transcriptionally regulate cellular fitness and functionality of T cell lineages through distinct gene programs. Immunobiology 2016; 221:618-33. [PMID: 26831822 DOI: 10.1016/j.imbio.2016.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 01/31/2023]
Abstract
T lymphocytes are essential contributors to the adaptive immune system and consist of multiple lineages that serve various effector and regulatory roles. As such, precise control of gene expression is essential to the proper development and function of these cells. Previously, we identified Snai2 and Snai3 as being essential regulators of immune tolerance partly due to the impaired function of CD4(+) regulatory T cells in Snai2/3 conditional double knockout mice. Here we extend those previous findings using a bone marrow transplantation model to provide an environmentally unbiased view of the molecular changes imparted onto various T lymphocyte populations once Snai2 and Snai3 are deleted. The data presented here demonstrate that Snai2 and Snai3 transcriptionally regulate the cellular fitness and functionality of not only CD4(+) regulatory T cells but effector CD8(α+) and CD4(+) conventional T cells as well. This is achieved through the modulation of gene sets unique to each cell type and includes transcriptional targets relevant to the survival and function of each T cell lineage. As such, Snai2 and Snai3 are essential regulators of T cell immunobiology.
Collapse
Affiliation(s)
- Peter D Pioli
- Division of Cell Biology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84132, United States.
| | - Sarah K Whiteside
- Division of Cell Biology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84132, United States
| | - Janis J Weis
- Division of Cell Biology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84132, United States
| | - John H Weis
- Division of Cell Biology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84132, United States
| |
Collapse
|