51
|
Wu F, Wang Z, Yang G, Jian J, Lu Y. Molecular characterization and expression analysis of interleukin-15 (IL-15) genes in orange-spotted grouper (Epinephelus coioides) in response to Vibrio harveyi challenge. FISH & SHELLFISH IMMUNOLOGY 2022; 128:327-334. [PMID: 35940540 DOI: 10.1016/j.fsi.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/24/2022] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
As a member of the γc family, interleukin 15 plays an important function in the immune response. In this study, we cloned an IL15 from Epinephelus coioides (named Ec-IL15). The open reading frame of Ec-IL15 is 528 bp, encoding 175 amino acids. Sequence alignment analysis showed that EcIL-15 has a conserved Pfam: IL15 domain and four cysteine residues. Subcellular localization studies have shown that Ec-IL15 is distributed in whole cells. In healthy groupers, Ec-IL15 was expressed in all 11 tissues tested and the highest in liver. After ConA, PHA, LPS and poly I:C stimulation, Ec-IL15 expression of HKLs was significantly upregulated. After V. harveyi infection, the expression of Ec-IL15 in 9 tissues was significantly upregulated and peaked within 48 h. In addition, recombinant Ec-IL15 protein can not only stimulate HKLs proliferation and cytokine expression, but also has the potential as an immune enhancer.
Collapse
Affiliation(s)
- Fan Wu
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 51820, China; College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Aquaic Animal Disease Control and Healthy Culture, Zhanjiang, 524025, China
| | - Zhiwen Wang
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 51820, China; College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Aquaic Animal Disease Control and Healthy Culture, Zhanjiang, 524025, China
| | - Guanjian Yang
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 51820, China; College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Aquaic Animal Disease Control and Healthy Culture, Zhanjiang, 524025, China
| | - Jichang Jian
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Aquaic Animal Disease Control and Healthy Culture, Zhanjiang, 524025, China
| | - Yishan Lu
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 51820, China; College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Aquaic Animal Disease Control and Healthy Culture, Zhanjiang, 524025, China.
| |
Collapse
|
52
|
The Critical Role of AMPKα1 in Regulating Autophagy and Mitochondrial Respiration in IL-15-Stimulated mTORC1Weak Signal-Induced T Cell Memory: An Interplay between Yin (AMPKα1) and Yang (mTORC1) Energy Sensors in T Cell Differentiation. Int J Mol Sci 2022; 23:ijms23179534. [PMID: 36076931 PMCID: PMC9455586 DOI: 10.3390/ijms23179534] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/29/2022] Open
Abstract
Two common γ-chain family cytokines IL-2 and IL-15 stimulate the same mammalian target of rapamycin complex-1 (mTORC1) signaling yet induce effector T (TE) and memory T (TM) cell differentiation via a poorly understood mechanism(s). Here, we prepared in vitro IL-2-stimulated TE (IL-2/TE) and IL-15-stimulated TM (IL-15/TM) cells for characterization by flow cytometry, Western blotting, confocal microscopy and Seahorse-assay analyses. We demonstrate that IL-2 and IL-15 stimulate strong and weak mTORC1 signals, respectively, which lead to the formation of CD62 ligand (CD62L)− killer cell lectin-like receptor subfamily G member-1 (KLRG)+ IL-2/TE and CD62L+KLRG− IL-15/TM cells with short- and long-term survival following their adoptive transfer into mice. The IL-15/mTORC1Weak signal activates the forkhead box-O-1 (FOXO1), T cell factor-1 (TCF1) and Eomes transcriptional network and the metabolic adenosine monophosphate-activated protein kinase-α-1 (AMPKα1), Unc-51-like autophagy-activating kinase-1 (ULK1) and autophagy-related gene-7 (ATG7) axis, increasing the expression of mitochondrial regulators aquaporin-9 (AQP9), mitochondrial transcription factor-A (TFAM), peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α), carnitine palmitoyl transferase-1 (CPT1α), microtubule-associated protein light chain-3 II (LC3II), Complex I and ortic atrophy-1 (OPA1), leading to promoting mitochondrial biogenesis and fatty-acid oxidation (FAO). Interestingly, AMPKα1 deficiency abrogates these downstream responses to IL-15/mTORC1Weak signaling, leading to the upregulation of mTORC1 and hypoxia-inducible factor-1α (HIF-1α), a metabolic switch from FAO to glycolysis and reduced cell survival. Taken together, our data demonstrate that IL-15/mTORC1Weak signaling controls T-cell memory via activation of the transcriptional FOXO1-TCF1-Eomes and metabolic AMPKα1-ULK1-ATG7 pathways, a finding that may greatly impact the development of efficient vaccines and immunotherapies for the treatment of cancer and infectious diseases.
Collapse
|
53
|
Carmenate T, Montalvo G, Lozada SL, Rodriguez Y, Ortiz Y, Díaz C, Avellanet J, Kim J, Surh CD, Graça L, León K. The antitumor effect induced by an IL-2 ‘no-alpha’ mutein depends on changes in the CD8+ T lymphocyte/Treg cell balance. Front Immunol 2022; 13:974188. [PMID: 36059465 PMCID: PMC9428827 DOI: 10.3389/fimmu.2022.974188] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/27/2022] [Indexed: 11/18/2022] Open
Abstract
High doses of interleukin-2 (IL-2) have been used for the treatment of melanoma and renal cell carcinoma, but this therapy has limited efficacy, with a ~15% response rate. Remarkably, 7%–9% of patients achieve complete or long-lasting responses. Many patients treated with IL-2 experienced an expansion of regulatory T cells (Tregs), specifically the expansion of ICOS+ highly suppressive Tregs, which correlate with worse clinical outcomes. This partial efficacy together with the high toxicity associated with the therapy has limited the use of IL-2-based therapy. Taking into account the understanding of IL-2 structure, signaling, and in vivo functions, some efforts to improve the cytokine properties are currently under study. In previous work, we described an IL-2 mutein with higher antitumor activity and less toxicity than wtIL-2. Mutein was in silico designed for losing the binding capacity to CD25 and for preferential stimulation of effector cells CD8+ and NK cells but not Tregs. Mutein induces a higher anti-metastatic effect than wtIL-2, but the extent of the in vivo antitumor activity was still unexplored. In this work, it is shown that mutein induces a strong antitumor effect on four primary tumor models, being effective even in those models where wtIL-2 does not work. Furthermore, mutein can change the in vivo balance between Tregs and T CD8+ memory/activated cells toward immune activation, in both healthy and tumor-bearing mice. This change reaches the tumor microenvironment and seems to be the major explanation for mutein efficacy in vivo.
Collapse
Affiliation(s)
- Tania Carmenate
- Immune Regulation Department, Centro de Inmunología Molecular, Havana, Cuba
- *Correspondence: Tania Carmenate,
| | - Galia Montalvo
- Immune Regulation Department, Centro de Inmunología Molecular, Havana, Cuba
| | - Sum Lai Lozada
- Immune Regulation Department, Centro de Inmunología Molecular, Havana, Cuba
| | - Yaretnis Rodriguez
- Immune Regulation Department, Centro de Inmunología Molecular, Havana, Cuba
| | - Yaquelin Ortiz
- Immune Regulation Department, Centro de Inmunología Molecular, Havana, Cuba
| | - Claudia Díaz
- Immune Regulation Department, Centro de Inmunología Molecular, Havana, Cuba
| | - Janet Avellanet
- Immune Regulation Department, Centro de Inmunología Molecular, Havana, Cuba
| | - Juhee Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science 12 and Technology (POSTECH), Pohang, South Korea
| | - Charles D. Surh
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science 12 and Technology (POSTECH), Pohang, South Korea
| | - Luis Graça
- Instituto de Medicina Molecular, Faculdade de Medici na da Universidade de Lisboa, Lisbon, Portugal
| | - Kalet León
- Immune Regulation Department, Centro de Inmunología Molecular, Havana, Cuba
| |
Collapse
|
54
|
Louie AY, Tingling J, Dray E, Hussain J, McKim DB, Swanson KS, Steelman AJ. Dietary Cholesterol Causes Inflammatory Imbalance and Exacerbates Morbidity in Mice Infected with Influenza A Virus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2523-2539. [PMID: 35577367 DOI: 10.4049/jimmunol.2100927] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 03/21/2022] [Indexed: 12/27/2022]
Abstract
Influenza is a common cause of pneumonia-induced hospitalization and death, but how host factors function to influence disease susceptibility or severity has not been fully elucidated. Cellular cholesterol levels may affect the pathogenesis of influenza infection, as cholesterol is crucial for viral entry and replication, as well as immune cell proliferation and function. However, there is still conflicting evidence on the extent to which dietary cholesterol influences cholesterol metabolism. In this study, we examined the effects of a high-cholesterol diet in modulating the immune response to influenza A virus (IAV) infection in mice. Mice were fed a standard or a high-cholesterol diet for 5 wk before inoculation with mouse-adapted human IAV (Puerto Rico/8/1934), and tissues were collected at days 0, 4, 8, and 16 postinfection. Cholesterol-fed mice exhibited dyslipidemia characterized by increased levels of total serum cholesterol prior to infection and decreased triglycerides postinfection. Cholesterol-fed mice also displayed increased morbidity compared with control-fed mice, which was neither a result of immunosuppression nor changes in viral load. Instead, transcriptomic analysis of the lungs revealed that dietary cholesterol caused upregulation of genes involved in viral-response pathways and leukocyte trafficking, which coincided with increased numbers of cytokine-producing CD4+ and CD8+ T cells and infiltrating dendritic cells. Morbidity as determined by percent weight loss was highly correlated with numbers of cytokine-producing CD4+ and CD8+ T cells as well as granulocytes. Taken together, dietary cholesterol promoted IAV morbidity via exaggerated cellular immune responses that were independent of viral load.
Collapse
Affiliation(s)
- Allison Y Louie
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Joseph Tingling
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Evan Dray
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Jamal Hussain
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Daniel B McKim
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL.,Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL; and
| | - Kelly S Swanson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL; and
| | - Andrew J Steelman
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL; .,Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL; and.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL
| |
Collapse
|
55
|
Jeong S, Jeon M, Lee H, Kim SY, Park SH, Shin EC. IFITM3 Is Upregulated Characteristically in IL-15-Mediated Bystander-Activated CD8 + T Cells during Influenza Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1901-1911. [PMID: 35346965 DOI: 10.4049/jimmunol.2100629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 02/07/2022] [Indexed: 06/14/2023]
Abstract
In bystander activation, pre-existing memory CD8+ T cells unrelated to the infecting microbes are activated by cytokines without cognate Ags. The detailed mechanisms and unique gene signature of bystander activation remain to be elucidated. In this study, we investigated bystander activation of OT-1 memory cells in a mouse model of influenza infection. We found that OT-1 memory cells are activated with upregulation of granzyme B and IFN-γ, during PR8 (A/Puerto Rico/8/1934) infection, and IL-15 is a critical cytokine for bystander activation. In transcriptomic analysis, the IFN-induced gene signature was upregulated in bystander-activated OT-1 memory cells during PR8 infection but not in the presence of TCR stimulation. Among the IFN-induced genes, upregulation of IFN-induced transmembrane protein 3 (IFITM3) distinguished bystander-activated OT-1 memory cells from TCR-activated OT-1 memory cells. Therefore, we reveal that bystander-activated memory CD8+ T cells have a unique transcriptomic feature compared with TCR-activated memory CD8+ T cells. In particular, IFITM3 upregulation can be used as a marker of bystander-activated memory CD8+ T cells at early infection.
Collapse
Affiliation(s)
- Seongju Jeong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Minwoo Jeon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Hoyoung Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - So-Young Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
56
|
Abstract
Autoimmune disease is known to be caused by unregulated self-antigen-specific T cells, causing tissue damage. Although antigen specificity is an important mechanism of the adaptive immune system, antigen non-related T cells have been found in the inflamed tissues in various conditions. Bystander T cell activation refers to the activation of T cells without antigen recognition. During an immune response to a pathogen, bystander activation of self-reactive T cells via inflammatory mediators such as cytokines can trigger autoimmune diseases. Other antigen-specific T cells can also be bystander-activated to induce innate immune response resulting in autoimmune disease pathogenesis along with self-antigen-specific T cells. In this review, we summarize previous studies investigating bystander activation of various T cell types (NKT, γδ T cells, MAIT cells, conventional CD4+, and CD8+ T cells) and discuss the role of innate-like T cell response in autoimmune diseases. In addition, we also review previous findings of bystander T cell function in infection and cancer. A better understanding of bystander-activated T cells versus antigen-stimulated T cells provides a novel insight to control autoimmune disease pathogenesis.
Collapse
Affiliation(s)
- Chae-Hyeon Shim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Sookyung Cho
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Young-Mi Shin
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Research Institute for Natural Sciences, Institute for Rheumatology Research, Research Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
57
|
Shim CH, Cho S, Shin YM, Choi JM. Emerging role of bystander T cell activation in autoimmune diseases. BMB Rep 2022; 55:57-64. [PMID: 35000675 PMCID: PMC8891623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/14/2021] [Accepted: 01/10/2022] [Indexed: 02/21/2025] Open
Abstract
Autoimmune disease is known to be caused by unregulated selfantigen-specific T cells, causing tissue damage. Although antigen specificity is an important mechanism of the adaptive immune system, antigen non-related T cells have been found in the inflamed tissues in various conditions. Bystander T cell activation refers to the activation of T cells without antigen recognition. During an immune response to a pathogen, bystander activation of self-reactive T cells via inflammatory mediators such as cytokines can trigger autoimmune diseases. Other antigen-specific T cells can also be bystander-activated to induce innate immune response resulting in autoimmune disease pathogenesis along with self-antigen-specific T cells. In this review, we summarize previous studies investigating bystander activation of various T cell types (NKT, γδ T cells, MAIT cells, conventional CD4+, and CD8+ T cells) and discuss the role of innate-like T cell response in autoimmune diseases. In addition, we also review previous findings of bystander T cell function in infection and cancer. A better understanding of bystander-activated T cells versus antigenstimulated T cells provides a novel insight to control autoimmune disease pathogenesis. [BMB Reports 2022; 55(2): 57-64].
Collapse
Affiliation(s)
- Chae-Hyeon Shim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Sookyung Cho
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Young-Mi Shin
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Research Institute for Natural Sciences, Institute for Rheumatology Research, Research Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
58
|
Kleinman AJ, Pandrea I, Apetrei C. So Pathogenic or So What?-A Brief Overview of SIV Pathogenesis with an Emphasis on Cure Research. Viruses 2022; 14:135. [PMID: 35062339 PMCID: PMC8781889 DOI: 10.3390/v14010135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/10/2021] [Accepted: 12/25/2021] [Indexed: 02/07/2023] Open
Abstract
HIV infection requires lifelong antiretroviral therapy (ART) to control disease progression. Although ART has greatly extended the life expectancy of persons living with HIV (PWH), PWH nonetheless suffer from an increase in AIDS-related and non-AIDS related comorbidities resulting from HIV pathogenesis. Thus, an HIV cure is imperative to improve the quality of life of PWH. In this review, we discuss the origins of various SIV strains utilized in cure and comorbidity research as well as their respective animal species used. We briefly detail the life cycle of HIV and describe the pathogenesis of HIV/SIV and the integral role of chronic immune activation and inflammation on disease progression and comorbidities, with comparisons between pathogenic infections and nonpathogenic infections that occur in natural hosts of SIVs. We further discuss the various HIV cure strategies being explored with an emphasis on immunological therapies and "shock and kill".
Collapse
Affiliation(s)
- Adam J. Kleinman
- Division of Infectious Diseases, DOM, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Ivona Pandrea
- Department of Infectious Diseases and Immunology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Cristian Apetrei
- Division of Infectious Diseases, DOM, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Department of Infectious Diseases and Immunology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| |
Collapse
|
59
|
Significance of bystander T cell activation in microbial infection. Nat Immunol 2022; 23:13-22. [PMID: 34354279 DOI: 10.1038/s41590-021-00985-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023]
Abstract
During microbial infection, pre-existing memory CD8+ T cells that are not specific for the infecting pathogens can be activated by cytokines without cognate antigens, termed bystander activation. Studies in mouse models and human patients demonstrate bystander activation of memory CD8+ T cells, which exerts either protective or detrimental effects on the host, depending on the infection model or disease. Research has elucidated mechanisms underlying the bystander activation of CD8+ T cells in terms of the responsible cytokines and the effector mechanisms of bystander-activated CD8+ T cells. In this Review, we describe the history of research on bystander CD8+ T cell activation as well as evidence of bystander activation. We also discuss the mechanisms and immunopathological roles of bystander activation in various microbial infections.
Collapse
|
60
|
Type I Interferon Promotes Antitumor T Cell Response in CRPC by Regulating MDSC. Cancers (Basel) 2021; 13:cancers13215574. [PMID: 34771735 PMCID: PMC8582786 DOI: 10.3390/cancers13215574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Despite initial tumor regression following androgen blockade treatment, relapse of castration-resistant prostate cancer (CRPC) eventually occurs in most patients. Immunotherapy aims to activate the host immune system to fight against cancer and has achieved significant therapeutic effects in various solid tumors. The purpose of our research was to investigate the mechanisms underlying the immune response during CRPC development and to screen effective immunotherapies against CRPC. We found that interferon-α (IFNα) directly inhibited the progression of CRPC, reduced the accumulation of the immune suppressive granulocytic myeloid-derived suppressor cells (G-MDSCs) in the tumor microenvironment (TME), and impaired the inhibitory function of G-MDSCs on T cell activation. This research provides a potential strategy for the clinical treatment of CRPC. Abstract Background: Metastatic castration-resistant prostate cancer (CRPC) is the leading cause of death among prostate cancer patients. Here, our aim was to ascertain the immune regulatory mechanisms involved in CRPC development and identify potential immunotherapies against CRPC. Methods: A CRPC model was established using Myc-CaP cells in immune-competent FVB mice following castration. The immune cell profile of the tumor microenvironment (TME) was analyzed during CRPC development. Different immunotherapies were screened in the CRPC tumor model, and their efficacies and underlying mechanisms were investigated in vitro and in vivo. Results: During CRPC development, the proportion of granulocytic myeloid-derived suppressor cells (G-MDSCs) in the TME increased. Among the immunotherapies tested, IFNα was more effective than anti-PD-L1, anti-CTLA-4, anti-4-1BB, IL-2, and IL-9 in reducing Myc-CaP CRPC tumor growth. IFNα reduced the number of G-MDSCs both in vitro during differentiation and in vivo in CRPC mice. Furthermore, IFNα reduced the suppressive function of G-MDSCs on T cell proliferation and activation. Conclusion: G-MDSCs are crucial to effective immunotherapy against CRPC. Treatment with IFNα presents a promising therapeutic strategy against CRPC. Besides the direct inhibition of tumor growth and the promotion of T cell priming, IFNα reduces the number and the suppressive function of G-MDSCs and restores T cell activation.
Collapse
|
61
|
Kosaka A, Ishibashi K, Nagato T, Kitamura H, Fujiwara Y, Yasuda S, Nagata M, Harabuchi S, Hayashi R, Yajima Y, Ohara K, Kumai T, Aoki N, Komohara Y, Oikawa K, Harabuchi Y, Kitada M, Kobayashi H, Ohkuri T. CD47 blockade enhances the efficacy of intratumoral STING-targeting therapy by activating phagocytes. J Exp Med 2021; 218:212661. [PMID: 34559187 PMCID: PMC8480673 DOI: 10.1084/jem.20200792] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/13/2021] [Accepted: 09/03/2021] [Indexed: 01/18/2023] Open
Abstract
Activation of STING signaling plays an important role in anti-tumor immunity, and we previously reported the anti-tumor effects of STING through accumulation of M1-like macrophages in tumor tissue treated with a STING agonist. However, myeloid cells express SIRPα, an inhibitory receptor for phagocytosis, and its receptor, CD47, is overexpressed in various cancer types. Based on our findings that breast cancer patients with highly expressed CD47 have poor survival, we evaluated the therapeutic efficacy and underlying mechanisms of combination therapy with the STING ligand cGAMP and an antagonistic anti-CD47 mAb using E0771 mouse breast cancer cells. Anti-CD47 mAb monotherapy did not suppress tumor growth in our setting, whereas cGAMP and anti-CD47 mAb combination therapy inhibited tumor growth. The combination therapy enhanced phagocytosis of tumor cells and induced systemic anti-tumor immune responses, which rely on STING and type I IFN signaling. Taken together, our findings indicate that coadministration of cGAMP and an antagonistic anti-CD47 mAb may be promising for effective cancer immunotherapy.
Collapse
Affiliation(s)
- Akemi Kosaka
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Kei Ishibashi
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.,Respiratory and Breast Center, Asahikawa Medical University Hospital, Asahikawa, Japan
| | - Toshihiro Nagato
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Hidemitsu Kitamura
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Syunsuke Yasuda
- Respiratory and Breast Center, Asahikawa Medical University Hospital, Asahikawa, Japan
| | - Marino Nagata
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Shohei Harabuchi
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.,Department of Otolaryngology, Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Ryusuke Hayashi
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.,Department of Otolaryngology, Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Yuki Yajima
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Kenzo Ohara
- Department of Otolaryngology, Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Takumi Kumai
- Department of Otolaryngology, Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Naoko Aoki
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kensuke Oikawa
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Yasuaki Harabuchi
- Department of Otolaryngology, Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Masahiro Kitada
- Respiratory and Breast Center, Asahikawa Medical University Hospital, Asahikawa, Japan
| | - Hiroya Kobayashi
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Takayuki Ohkuri
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
62
|
Li G, Zhang Z, Cai L, Tang X, Huang J, Yu L, Wang G, Zhong K, Cao Y, Liu C, Wang Y, Tong A, Zhou L. Fn14-targeted BiTE and CAR-T cells demonstrate potent preclinical activity against glioblastoma. Oncoimmunology 2021; 10:1983306. [PMID: 34595061 PMCID: PMC8477963 DOI: 10.1080/2162402x.2021.1983306] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
T cell-engaging therapies involving bispecific T cell engager (BiTE) and chimeric antigen receptor T (CAR-T) cells have achieved great success in the treatment of hematological tumors. However, the paucity of ideal cell surface molecules that can be targeted on glioblastoma (GBM) partially reduces the immunotherapeutic efficacy. Recently, high expression of Fn14 has been reported in several solid tumors, so the strategy of exploiting this specific antigen for GBM immunotherapy is worth studying. Consequently, we constructed Fn14× CD3 BiTE and Fn14-specific CAR-T cells and investigated their cytotoxic activity against GBM in vitro and in vivo. First, expression of Fn14 was confirmed in glioma tissues and GBM cells. Then, we designed Fn14-specific BiTE and CAR-T cells and tested their cytotoxicity in GBM cell cultures and mouse models of GBM. Fn14 was highly expressed in GBM tissues and cell lines, while it was undetectable in normal brain samples. Fn14× CD3 BiTE, Fn14 CAR-T cells and Fn14 CAR-T/IL-15 cells were antigen-specific and highly cytotoxic, showing good antitumor activity in vitro and causing significant regression of established solid tumors in xenograft models. However, the xenografts treated with Fn14 CAR-T cells regrew, whereas xenografts treated with Fn14 CAR-T/IL-15 cells did not. IL-15 engineering augmented the antitumor activity of Fn14 CAR-T cells and resulted in significant antitumor effects similar to those of Fn14× CD3 BiTE. Our results suggest that Fn14 is an appropriate target for GBM. Anti-Fn14 BiTE and Fn14-specific CAR-T/IL-15 cells may be exciting immunotherapeutic options for malignant brain cancer.
Collapse
Affiliation(s)
- Gaowei Li
- Department of Neurosurgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Zongliang Zhang
- State Key Laboratory of Biotherapy, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Linjun Cai
- Department of Neurology, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Xin Tang
- Department of Neurosurgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Jianhan Huang
- Department of Neurosurgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Lingyu Yu
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Guoqing Wang
- Department of Neurosurgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Kunhong Zhong
- State Key Laboratory of Biotherapy, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Yi Cao
- Department of Neurosurgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Chang Liu
- Department of Neurosurgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Yuelong Wang
- Department of Neurosurgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Aiping Tong
- State Key Laboratory of Biotherapy, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Liangxue Zhou
- Department of Neurosurgery, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| |
Collapse
|
63
|
Functional NK Cell Activation by Ovalbumin Immunization with a Monophosphoryl Lipid A and Poly I:C Combination Adjuvant Promoted Dendritic Cell Maturation. Vaccines (Basel) 2021; 9:vaccines9101061. [PMID: 34696169 PMCID: PMC8540815 DOI: 10.3390/vaccines9101061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/19/2021] [Accepted: 09/19/2021] [Indexed: 12/02/2022] Open
Abstract
Natural killer (NK) cells are one of the types of innate immune cells to remove pathogen-infected cells and modulate inflammatory immune responses. Recent studies have revealed that NK cells could enhance vaccine efficacy by coordinating the innate and adaptive immune responses. In this study, we have evaluated the efficacy of intranasal ovalbumin (OVA) immunization with a monophosphoryl lipid A (MPL) and polyriboinosinic polyribocytidylic acid (poly I:C) combination adjuvant in promoting NK cell recruitment, differentiation, and activation. The frequencies of NK cells were positively correlated with those of dendritic cells (DCs) at the site of immunization. Moreover, the activated NK cells and DCs by the MPL + poly I:C combination adjuvant induced activations of each other cells in vitro. Taken together, this study suggested that the MPL and poly I:C combination adjuvant in OVA vaccination mediated NK cell activation and cellular crosstalk between NK cells and DCs, suggesting a promising vaccine adjuvant candidate for promoting cellular immune responses.
Collapse
|
64
|
Bosch NC, Martin LM, Voskens CJ, Berking C, Seliger B, Schuler G, Schaft N, Dörrie J. A Chimeric IL-15/IL-15Rα Molecule Expressed on NFκB-Activated Dendritic Cells Supports Their Capability to Activate Natural Killer Cells. Int J Mol Sci 2021; 22:ijms221910227. [PMID: 34638566 PMCID: PMC8508776 DOI: 10.3390/ijms221910227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/02/2021] [Accepted: 09/18/2021] [Indexed: 12/31/2022] Open
Abstract
Natural killer (NK) cells, members of the innate immune system, play an important role in the rejection of HLA class I negative tumor cells. Hence, a therapeutic vaccine, which can activate NK cells in addition to cells of the adaptive immune system might induce a more comprehensive cellular response, which could lead to increased tumor elimination. Dendritic cells (DCs) are capable of activating and expanding NK cells, especially when the NFκB pathway is activated in the DCs thereby leading to the secretion of the cytokine IL-12. Another prominent NK cell activator is IL-15, which can be bound by the IL-15 receptor alpha-chain (IL-15Rα) to be transpresented to the NK cells. However, monocyte-derived DCs do neither secrete IL-15, nor express the IL-15Rα. Hence, we designed a chimeric protein consisting of IL-15 and the IL-15Rα. Upon mRNA electroporation, the fusion protein was detectable on the surface of the DCs, and increased the potential of NFκB-activated, IL-12-producing DC to activate NK cells in an autologous cell culture system with ex vivo-generated cells from healthy donors. These data show that a chimeric IL-15/IL-15Rα molecule can be expressed by monocyte-derived DCs, is trafficked to the cell surface, and is functional regarding the activation of NK cells. These data represent an initial proof-of-concept for an additional possibility of further improving cellular DC-based immunotherapies of cancer.
Collapse
Affiliation(s)
- Naomi C. Bosch
- Institute of Medical Immunology, Martin-Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (N.C.B.); (B.S.)
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
- Comprehensive Cancer Center Erlangen–EMN, NCT WERA, 91054 Erlangen, Germany
| | - Lena-Marie Martin
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
| | - Caroline J. Voskens
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
- Comprehensive Cancer Center Erlangen–EMN, NCT WERA, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
- Comprehensive Cancer Center Erlangen–EMN, NCT WERA, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin-Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (N.C.B.); (B.S.)
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), 04103 Leipzig, Germany
| | - Gerold Schuler
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
| | - Niels Schaft
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
| | - Jan Dörrie
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.-M.M.); (C.J.V.); (C.B.); (G.S.); (N.S.)
- Correspondence: ; Tel.: +49-9131-8531127
| |
Collapse
|
65
|
Jia X, Chua BY, Loh L, Koutsakos M, Kedzierski L, Olshansky M, Heath WR, Chang SY, Xu J, Wang Z, Kedzierska K. High expression of CD38 and MHC class II on CD8 + T cells during severe influenza disease reflects bystander activation and trogocytosis. Clin Transl Immunology 2021; 10:e1336. [PMID: 34522380 PMCID: PMC8426257 DOI: 10.1002/cti2.1336] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/19/2021] [Accepted: 08/10/2021] [Indexed: 11/12/2022] Open
Abstract
Objectives Although co‐expression of CD38 and HLA‐DR reflects T‐cell activation during viral infections, high and prolonged CD38+HLA‐DR+ expression is associated with severe disease. To date, the mechanism underpinning expression of CD38+HLA‐DR+ is poorly understood. Methods We used mouse models of influenza A/H9N2, A/H7N9 and A/H3N2 infection to investigate mechanisms underpinning CD38+MHC‐II+ phenotype on CD8+ T cells. To further understand MHC‐II trogocytosis on murine CD8+ T cells as well as the significance behind the scenario, we used adoptively transferred transgenic OT‐I CD8+ T cells and A/H3N2‐SIINKEKL infection. Results Analysis of influenza‐specific immunodominant DbNP366+CD8+ T‐cell responses showed that CD38+MHC‐II+ co‐expression was detected on both virus‐specific and bystander CD8+ T cells, with increased numbers of both CD38+MHC‐II+CD8+ T‐cell populations observed in immune organs including the site of infection during severe viral challenge. OT‐I cells adoptively transferred into MHC‐II−/− mice had no MHC‐II after infection, suggesting that MHC‐II was acquired via trogocytosis. The detection of CD19 on CD38+MHC‐II+ OT‐I cells supports the proposition that MHC‐II was acquired by trogocytosis sourced from B cells. Co‐expression of CD38+MHC‐II+ on CD8+ T cells was needed for optimal recall following secondary infection. Conclusions Overall, our study demonstrates that both virus‐specific and bystander CD38+MHC‐II+ CD8+ T cells are recruited to the site of infection during severe disease, and that MHC‐II presence occurs via trogocytosis from antigen‐presenting cells. Our findings highlight the importance of the CD38+MHC‐II+ phenotype for CD8+ T‐cell recall.
Collapse
Affiliation(s)
- Xiaoxiao Jia
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - Brendon Y Chua
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - Liyen Loh
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia.,Faculty of Veterinary and Agricultural Sciences University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - Moshe Olshansky
- Department of Microbiology Monash University Clayton VIC Australia
| | - William R Heath
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - So Young Chang
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| | - Jianqing Xu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences Key Laboratory of Medical Molecular Virology of Ministry of Education/Health Shanghai Medical College Fudan University Shanghai China
| | - Zhongfang Wang
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia.,State Key Laboratory of Respiratory Disease Guangzhou Medical University Guangzhou China
| | - Katherine Kedzierska
- Department of Microbiology and Immunology University of Melbourne, at the Peter Doherty Institute for Infection and Immunity Parkville VIC Australia
| |
Collapse
|
66
|
Devalraju KP, Neela VSK, Krovvidi SS, Vankayalapati R, Valluri VL. Defective expansion and function of memory like natural killer cells in HIV+ individuals with latent tuberculosis infection. PLoS One 2021; 16:e0257185. [PMID: 34516566 PMCID: PMC8437280 DOI: 10.1371/journal.pone.0257185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 08/25/2021] [Indexed: 11/29/2022] Open
Abstract
PURPOSE Tuberculosis (TB) is the leading cause of infectious disease related mortality, and only 10% of the infected individuals develop active disease. The likelihood of progression of latent tuberculosis infection (LTBI) to active TB disease is high in HIV infected individuals. Identification of HIV+ individuals at risk would allow treating targeted population, facilitating completion of therapy for LTBI and prevention of TB development. NK cells have an important role in T cell independent immunity against TB, but the exact role of NK cell subsets in LTBI and HIV is not well characterized. METHODS In this study, we compared the expansion and function of memory like NK cells from HIV-LTBI+ individuals and treatment naïve HIV+LTBI+ patients in response to Mtb antigens ESAT-6 and CFP-10. RESULTS In freshly isolated PBMCs, percentages of CD3-CD56+ NK cells were similar in HIV+LTBI+ patients and healthy HIV-LTBI+ individuals. However, percentages of CD3-CD56+CD16+ NK cells were higher in healthy HIV-LTBI+ individuals compared to HIV+LTBI+ patients. HIV infection also inhibited the expansion of memory like NK cells, production of IL-32α, IL-15 and IFN-γ in response to Mtb antigens in LTBI+ individuals. CONCLUSION We studied phenotypic, functional subsets and activation of memory like-NK cells during HIV infection and LTBI. We observed that HIV+LTBI+ patients demonstrated suboptimal NK cell and monocyte interactions in response to Mtb, leading to reduced IL-15, IFN-γ and granzyme B and increased CCL5 production. Our study highlights the effect of HIV and LTBI on modulation of NK cell activity to understand their role in development of interventions to prevent progression to TB in high risk individuals.
Collapse
Affiliation(s)
- Kamakshi Prudhula Devalraju
- Immunology and Molecular Biology Division, Blue Peter Public Health and Research Centre, LEPRA Society, Cherlapally, Hyderabad, Telangana, India
| | - Venkata Sanjeev Kumar Neela
- Immunology and Molecular Biology Division, Blue Peter Public Health and Research Centre, LEPRA Society, Cherlapally, Hyderabad, Telangana, India
| | - Siva Sai Krovvidi
- Department of Biotechnology, Sreenidhi Institute of Science and Technology, Yamnampet, Ghatkesar, Hyderabad, Telangana, India
| | - Ramakrishna Vankayalapati
- Department of Pulmonary Immunology, Center for Biomedical Research, The University of Texas Health Center at Tyler, Texas, TX, United States of America
| | - Vijaya Lakshmi Valluri
- Immunology and Molecular Biology Division, Blue Peter Public Health and Research Centre, LEPRA Society, Cherlapally, Hyderabad, Telangana, India
| |
Collapse
|
67
|
Hsieh EW, Hernandez JD. Clean up by aisle 2: roles for IL-2 receptors in host defense and tolerance. Curr Opin Immunol 2021; 72:298-308. [PMID: 34479098 DOI: 10.1016/j.coi.2021.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 12/24/2022]
Abstract
Although IL-2 was first recognized as growth factor for T cells, it is now also appreciated to be a key regulator of T cells through its effects on regulatory T cells (Treg). The IL-2 receptor (IL-2R) subunits' different (i) ligand affinities, (ii) dimerization or trimerization relationships with other cytokine subunits, (iii) expression across multiple cell types, and (iv) downstream signaling effects, largely dictate cellular tolerance and antimicrobial processes. Defects in IL-2Rγ result in profound and almost universally fatal immune deficiency, unless treated with hematopoietic stem cell transplantation (HSCT). Defects in IL-2Rα and IL-2Rβ result in more limited infection susceptibility, particularly to herpesviruses. However, the most prominent clinical symptomatology for IL-2Rα and IL-2Rβ defects include multi-organ autoimmunity and inflammation, consistent with the critical role of IL-2 in establishing and maintaining immune tolerance. Here, we review how we have arrived at our current understanding of the complex roles of IL-2/2R in host defense and tolerance focusing on the insights gained from human clinical immunology.
Collapse
Affiliation(s)
- Elena Wy Hsieh
- Department of Pediatrics, Section of Allergy and Immunology, School of Medicine, University of Colorado, Children's Hospital Colorado, United States; Department of Immunology and Microbiology, School of Medicine, University of Colorado, United States.
| | - Joseph D Hernandez
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, School of Medicine, Stanford University, Lucile Packard Children's Hospital, United States
| |
Collapse
|
68
|
Abstract
Conventional CD4+ and CD8+ T lymphocytes comprise a mixture of naive and memory cells. Generation and survival of these T-cell subsets is under strict homeostatic control and reflects contact with self-major histocompatibility complex (MHC) and certain cytokines. Naive T cells arise in the thymus via T-cell receptor (TCR)-dependent positive selection to self-peptide/MHC complexes and are then maintained in the periphery through self-MHC interaction plus stimulation via interleukin-7 (IL-7). By contrast, memory T cells are largely MHC-independent for their survival but depend strongly on stimulation via cytokines. Whereas typical memory T cells are generated in response to foreign antigens, some arise spontaneously through contact of naive precursors with self-MHC ligands; we refer to these cells as memory-phenotype (MP) T cells. In this review, we discuss the generation and homeostasis of naive T cells and these two types of memory T cells, focusing on their relative interaction with MHC ligands and cytokines.
Collapse
Affiliation(s)
- Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Jaeu Yi
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Jonathan Sprent
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| |
Collapse
|
69
|
McLeod IX, Saxena R, Carico Z, He YW. Class I PI3K Provide Lipid Substrate in T Cell Autophagy Through Linked Activity of Inositol Phosphatases. Front Cell Dev Biol 2021; 9:709398. [PMID: 34458267 PMCID: PMC8397451 DOI: 10.3389/fcell.2021.709398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/21/2021] [Indexed: 11/13/2022] Open
Abstract
Autophagy, a highly conserved intracellular process, has been identified as a novel mechanism regulating T lymphocyte homeostasis. Herein, we demonstrate that both starvation- and T cell receptor-mediated autophagy induction requires class I phosphatidylinositol-3 kinases to produce PI(3)P. In contrast, common gamma chain cytokines are suppressors of autophagy despite their ability to activate the PI3K pathway. T cells lacking the PI3KI regulatory subunits, p85 and p55, were almost completely unable to activate TCR-mediated autophagy and had concurrent defects in PI(3)P production. Additionally, T lymphocytes upregulate polyinositol phosphatases in response to autophagic stimuli, and the activity of the inositol phosphatases Inpp4 and SHIP are required for TCR-mediated autophagy induction. Addition of exogenous PI(3,4)P2 can supplement cellular PI(3)P and accelerate the outcome of activation-induced autophagy. TCR-mediated autophagy also requires internalization of the TCR complex, suggesting that this kinase/phosphatase activity is localized in internalized vesicles. Finally, HIV-induced bystander CD4+ T cell autophagy is dependent upon PI3KI. Overall, our data elucidate an important pathway linking TCR activation to autophagy, via induction of PI3KI activity and inositol phosphatase upregulation to produce PI(3)P.
Collapse
Affiliation(s)
- Ian X McLeod
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| | - Ruchi Saxena
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| | - Zachary Carico
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| | - You-Wen He
- Department of Immunology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
70
|
Robinson TO, Hegde SM, Chang AJ, Gangadharan A, Rivas S, Madakamutil L, Zalevsky J, Miyazaki T, Schluns KS. NKTR-255, a polymer-conjugated IL-15 with unique mechanisms of action on T and natural killer cells. J Clin Invest 2021; 131:e144365. [PMID: 34375310 DOI: 10.1172/jci144365] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 08/06/2021] [Indexed: 11/17/2022] Open
Abstract
NKTR-255 is a novel polyethylene glycol (PEG)-conjugate of recombinant human IL-15 (rhIL-15) being examined as a potential cancer immunotherapeutic. Since IL-15 responses can be mediated by trans- or cis-presentation via IL-15Rα or soluble IL-15/IL-15Rα complexes, we investigated the role of IL-15Rα in driving NKTR-255 responses using defined naïve and memory ovalbumin-specific CD8 T cells (OT-I) CD8 T and NK cells in mice. NKTR-255 induced a 2.5 and 2.0-fold expansion of CD8 T and NK cells, respectively in WT mice. In adoptive transfer studies, proliferation of naïve and memory Wt OT-I T cells in response to NKTR-255 was not impaired in IL-15Rα-/- mice, suggesting trans-presentation was not utilized by NKTR-255. Interestingly, naïve IL-15Rα-/- OT-I cells had deficient responses to NKTR-255 while memory IL-15Rα-/- OT-I cell responses were partially impaired, suggesting that naive CD8 T cells are more dependent on cis-presentation of NKTR-255 than memory CD8 T cells. In bone marrow chimeras studies, IL-15Rα-/- and WT NK cells present in WT recipients had similar responses to NKTR-255, suggesting that cis-presentation is not utilized by NK cells. NKTR-255 could form soluble complexes with IL-15Rα; binding to murine IL-15Rα generated superagonists that preferentially stimulated NK cells showing that conversion to IL-15Rβ agonist biases the response towards NK cells. These findings highlight the ability of NKTR-255 to utilize IL-15Rα for cis-presentation and act as an IL-15Rαβ agonist on CD8 T cells.
Collapse
Affiliation(s)
- Tanya O Robinson
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Shweta M Hegde
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Allison J Chang
- Chemical and Biomedical Engineering, Rice University, Houston, United States of America
| | - Achintyan Gangadharan
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Sarai Rivas
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Loui Madakamutil
- Research Biology: Pharmacology, Nektar Therapeutics, San Francisco, United States of America
| | - Jonathan Zalevsky
- Research and Development, Nektar Therapeutics, San Francisco, United States of America
| | - Takahiro Miyazaki
- Research Biology: Pharmacology, Nektar Therapeutics, San Francisco, United States of America
| | - Kimberly S Schluns
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, United States of America
| |
Collapse
|
71
|
Méndez-Lagares G, Chin N, Chang WW, Lee J, Rosás-Umbert M, Kieu HT, Merriam D, Lu W, Kim S, Adamson L, Brander C, Luciw PA, Barry PA, Hartigan-O’Connor DJ. Cytomegalovirus mediates expansion of IL-15-responsive innate-memory cells with SIV killing function. J Clin Invest 2021; 131:148542. [PMID: 34153005 PMCID: PMC8321572 DOI: 10.1172/jci148542] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
Interindividual immune variability is driven predominantly by environmental factors, including exposure to chronic infectious agents such as cytomegalovirus (CMV). We investigated the effects of rhesus CMV (RhCMV) on composition and function of the immune system in young macaques. Within months of infection, RhCMV was associated with impressive changes in antigen presenting cells, T cells, and NK cells-and marked expansion of innate-memory CD8+ T cells. These cells express high levels of NKG2A/C and the IL-2 and IL-15 receptor beta chain, CD122. IL-15 was sufficient to drive differentiation of the cells in vitro and in vivo. Expanded NKG2A/C+CD122+CD8+ T cells in RhCMV-infected macaques, but not their NKG2-negative counterparts, were endowed with cytotoxicity against class I-deficient K562 targets and prompt IFN-γ production in response to stimulation with IL-12 and IL-18. Because RhCMV clone 68-1 forms the viral backbone of RhCMV-vectored SIV vaccines, we also investigated immune changes following administration of RhCMV 68-1-vectored SIV vaccines. These vaccines led to impressive expansion of NKG2A/C+CD8+ T cells with capacity to inhibit SIV replication ex vivo. Thus, CMV infection and CMV-vectored vaccination drive expansion of functional innate-like CD8 cells via host IL-15 production, suggesting that innate-memory expansion could be achieved by other vaccine platforms expressing IL-15.
Collapse
Affiliation(s)
- Gema Méndez-Lagares
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - Ning Chin
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - W.L. William Chang
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - Jaewon Lee
- Graduate Group in Immunology, and
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | | | - Hung T. Kieu
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - David Merriam
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - Wenze Lu
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - Sungjin Kim
- Department of Medical Microbiology and Immunology
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Lourdes Adamson
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Christian Brander
- IrsiCaixa - AIDS Research Institute, Badalona, Barcelona, Spain
- Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Vic, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Paul A. Luciw
- California National Primate Research Center
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
- Department of Pathology and Laboratory Medicine, University of California, Davis, California, USA
| | - Peter A. Barry
- California National Primate Research Center
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
- Department of Pathology and Laboratory Medicine, University of California, Davis, California, USA
| | - Dennis J. Hartigan-O’Connor
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
72
|
Systemic IL-15, IFN-γ, and IP-10/CXCL10 signature associated with effective immune response to SARS-CoV-2 in BNT162b2 mRNA vaccine recipients. Cell Rep 2021; 36:109504. [PMID: 34352226 PMCID: PMC8299183 DOI: 10.1016/j.celrep.2021.109504] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/16/2021] [Accepted: 07/19/2021] [Indexed: 12/29/2022] Open
Abstract
Early responses to vaccination are important for shaping both humoral and cellular protective immunity. Dissecting innate vaccine signatures may predict immunogenicity to help optimize the efficacy of mRNA and other vaccine strategies. Here, we characterize the cytokine and chemokine responses to the 1st and 2nd dose of the BNT162b2 mRNA (Pfizer/BioNtech) vaccine in antigen-naive and in previously coronavirus disease 2019 (COVID-19)-infected individuals (NCT04743388). Transient increases in interleukin-15 (IL-15) and interferon gamma (IFN-γ) levels early after boost correlate with Spike antibody levels, supporting their use as biomarkers of effective humoral immunity development in response to vaccination. We identify a systemic signature including increases in IL-15, IFN-γ, and IP-10/CXCL10 after the 1st vaccination, which were enriched by tumor necrosis factor alpha (TNF-α) and IL-6 after the 2nd vaccination. In previously COVID-19-infected individuals, a single vaccination results in both strong cytokine induction and antibody titers similar to the ones observed upon booster vaccination in antigen-naive individuals, a result with potential implication for future public health recommendations.
Collapse
|
73
|
Abbas AA, Akbar AN. Induction of T Cell Senescence by Cytokine Induced Bystander Activation. FRONTIERS IN AGING 2021; 2:714239. [PMID: 35821998 PMCID: PMC9261416 DOI: 10.3389/fragi.2021.714239] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022]
Abstract
As people around the world continue to live longer, maintaining a good quality of life is of increasing importance. The COVID-19 pandemic revealed that the elderly are disproportionally vulnerable to infectious diseases and Immunosenescence plays a critical role in that. An ageing immune system influences the conventional activity of T cells which are at the forefront of eliminating harmful foreign antigens. With ageing, unconventional end-stage T cells, that exhibit a senescent phenotype, amass. These senescent T cells deviate from T cell receptor (TCR) signaling toward natural killer (NK) activity. The transition toward innate immune cell function from these adaptor T cells impacts antigen specificity, contributing to increased susceptibility of infection in the elderly. The mechanism by which senescent T cells arise remains largely unclear however in this review we investigate the part that bystander activation plays in driving the change in function of T cells with age. Cytokine-induced bystander activation may offer a plausible explanation for the induction of NK-like activity and senescence in T cells. Further understanding of these specific NK-like senescent T cells allows us to identify the benefits and detriments of these cells in health and disease which can be utilized or regulated, respectively. This review discusses the dynamic of senescent T cells in adopting NK-like T cells and the implications that has in an infectious disease context, predominately in the elderly.
Collapse
|
74
|
Stamova S, Ott-Rötzer B, Smetak H, Schäffler K, Eder R, Fink I, Hoffmann P, Reichert TE, Beckhove P, Spanier G. Characterization and ex vivo expansion of rare in situ cytokine secreting T cell populations from tumor tissue and blood of oral squamous cell carcinoma patients. J Immunol Methods 2021; 496:113086. [PMID: 34146580 DOI: 10.1016/j.jim.2021.113086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/14/2021] [Accepted: 06/14/2021] [Indexed: 12/18/2022]
Abstract
Rare subpopulations of tumor antigen-reactive memory T cells, which actively secrete type-1 effector cytokines, particularly TNF-α in situ, possess anti-tumor activity and prognostic relevance. These cells are relevant for cancer immunotherapy; however, their low frequencies make them difficult to study and novel protocols for their culture and expansion ex vivo are needed. Here, we studied the presence of T cells secreting type-1 cytokines (Cy+T cells) in the blood and tumors of 24 patients with oral squamous cell carcinomas (OSCC) and explored possibilities for their isolation and expansion. More than 90% of OSCC patients contained enriched numbers Cy+T cells in the blood and tumors compared to healthy donors in which these were hardly detectable. The majority of TNF-α+T cells were CD4+ T helper cells while IFN-γ+TIL were predominantly CD8+. Cy+T helper cells in the blood were early-differentiated memory T cells while Cy+TIL and Cy+CD8+T cells showed advanced-differentiated memory T cell phenotypes. We explored different conditions for their in vitro culture and found that Cy+T cells can be efficiently expanded in vitro to similar levels as Cy-T cells and after expansion maintained their TNF-α secreting capacity. However, for optimal expansion they required specific culture conditions to support the maintenance of stem-like and central memory T cell phenotype. In conclusion, we show that Cy+T cells are enriched in OSCC patients and report a novel cell culture protocol optimized to specifically expand and functionally maintain these cells for further functional characterization or for their exploitation in immunotherapy of OSCC.
Collapse
Affiliation(s)
- Slava Stamova
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany
| | - Birgitta Ott-Rötzer
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany
| | - Heiko Smetak
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany
| | - Katharina Schäffler
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany
| | - Rüdiger Eder
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Irina Fink
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany
| | - Petra Hoffmann
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany; Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Torsten E Reichert
- Department of Cranio-Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Philipp Beckhove
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany; Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany.
| | - Gerrit Spanier
- Department of Cranio-Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
75
|
Diversity, localization, and (patho)physiology of mature lymphocyte populations in the bone marrow. Blood 2021; 137:3015-3026. [PMID: 33684935 DOI: 10.1182/blood.2020007592] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/25/2021] [Indexed: 02/07/2023] Open
Abstract
The bone marrow (BM) is responsible for generating and maintaining lifelong output of blood and immune cells. In addition to its key hematopoietic function, the BM acts as an important lymphoid organ, hosting a large variety of mature lymphocyte populations, including B cells, T cells, natural killer T cells, and innate lymphoid cells. Many of these cell types are thought to visit the BM only transiently, but for others, like plasma cells and memory T cells, the BM provides supportive niches that promote their long-term survival. Interestingly, accumulating evidence points toward an important role for mature lymphocytes in the regulation of hematopoietic stem cells (HSCs) and hematopoiesis in health and disease. In this review, we describe the diversity, migration, localization, and function of mature lymphocyte populations in murine and human BM, focusing on their role in immunity and hematopoiesis. We also address how various BM lymphocyte subsets contribute to the development of aplastic anemia and immune thrombocytopenia, illustrating the complexity of these BM disorders and the underlying similarities and differences in their disease pathophysiology. Finally, we summarize the interactions between mature lymphocytes and BM resident cells in HSC transplantation and graft-versus-host disease. A better understanding of the mechanisms by which mature lymphocyte populations regulate BM function will likely improve future therapies for patients with benign and malignant hematologic disorders.
Collapse
|
76
|
Mulholland M, Jakobsson G, Lei Y, Sundius L, Ljungcrantz I, Rattik S, Tietge UJF, Engelbertsen D. IL-2Rβγ signalling in lymphocytes promotes systemic inflammation and reduces plasma cholesterol in atherosclerotic mice. Atherosclerosis 2021; 326:1-10. [PMID: 33945906 DOI: 10.1016/j.atherosclerosis.2021.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/08/2021] [Accepted: 04/21/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS The relationship between inflammation and lipid metabolism is complex and bidirectional. Lymphocyte-driven inflammation has been shown to modulate both atherosclerotic plaque development and cholesterol levels, but the mechanisms are incompletely understood. METHODS The cardiometabolic effects of IL-2Rβγ signalling in atherosclerotic Apoe-/- mice were investigated by treatment with an agonistic IL-2Rβγ-targeting IL-2/anti-IL-2 complex or a monoclonal anti-CD122 (IL-2Rβ) blocking antibody. RESULTS Administration of IL-2Rβγ agonistic IL-2/anti-IL-2 complexes to Apoe-/- mice augmented opposing arms of the adaptive immune system. Expansion of effector/memory T cells and increased levels of circulating pro-inflammatory cytokines were observed along with elevated levels of regulatory T cells and IL-10. Notably, IL-2/anti-IL-2 treatment did not affect plaque size but decreased levels of plasma cholesterol. The cholesterol lowering effect of IL-2Rβγ agonism was not affected by anti-CD8 or anti-NK1.1 depleting antibody treatment but was contingent on the presence of adaptive immunity. Expression of multiple liver X receptor (LXR)-related genes, including Pltp and Srebp1c in the liver, was decreased by IL-2/anti-IL-2 treatment. Although IL-2Rβγ agonism lowered cholesterol levels, blocking IL-2Rβγ signalling using an anti-CD122 monoclonal antibody did not impact cholesterol levels or plaque burden in Apoe-/- mice. CONCLUSIONS Elevated IL-2Rβγ signalling results in activation of both inflammatory and regulatory lymphocytes with a net zero effect on atherosclerosis and decreased plasma cholesterol levels. Changes in cholesterol levels were associated with reductions in hepatic LXR-related gene expression. Further studies are needed to investigate the clinical significance of IL-2 mediated modulation of hepatic LXR signalling in inflammatory disorders.
Collapse
Affiliation(s)
- Megan Mulholland
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden.
| | - Gabriel Jakobsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden
| | - Yu Lei
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, S-14183, Stockholm, Sweden
| | - Lena Sundius
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden
| | - Irena Ljungcrantz
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden
| | - Sara Rattik
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden
| | - Uwe J F Tietge
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, S-14183, Stockholm, Sweden; Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, SE-141 86, Stockholm, Sweden
| | - Daniel Engelbertsen
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden.
| |
Collapse
|
77
|
Tarantino G, Citro V, Balsano C, Capone D. Age and Interleukin-15 Levels Are Independently Associated With Intima-Media Thickness in Obesity-Related NAFLD Patients. Front Med (Lausanne) 2021; 8:634962. [PMID: 34095164 PMCID: PMC8175965 DOI: 10.3389/fmed.2021.634962] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 04/06/2021] [Indexed: 01/06/2023] Open
Abstract
Common carotid intima-media thickness (IMT) represents a functional and structural marker of early, precocious, and subclinical atherosclerosis, independently from the carotid plaque. Macrophage cells, which have been detected in adipose tissue and atherosclerotic plaques, are regulated by interleukin-15 (IL-15). At the light of the conflicting results concerning the role of IL-15 in atherosclerosis, the aim of the study was to retrospectively evaluate in a population of 80 obese patients, with median age of 46 years (IQR 34-53 years), with a low rate of comorbidities but with non-alcoholic fatty liver disease (NAFLD) or hepatic steatosis (HS), the relationship between IMT and serum concentrations of IL-15. Anthropometric measures, metabolic profile, and serum inflammatory markers, as well as the levels of IL-15, MCP-1, b FGF, and GM-CSF, were analyzed by a bead-based assay. IMT, HS, subcutaneous, and visceral adipose tissues were detected by ultrasonography. The IL-15 levels of the obese patients were increased with respect to those of 44 young healthy subjects, i.e., 2.77 (1.21-4.8) vs. 1.55 (1-2.4) pg/mL (P = 0.002). In the univariate analysis, IL-15 levels were associated to IMT and to those of MCP-1, b FGF, and GM-CSF, without any relation to other inflammatory markers such as CRP and ferritin, except fibrinogen. In the multivariate analysis, after adjusting the HS severity for the extent of visceral adiposity, a dramatic change in prediction of IMT by HS was shown (β from 0.29 to 0.10, P from 0.008 to 0.37). When the visceral adipose tissue was combined with IL-15, on the one hand, and the well-known coronary artery disease (CAD) risk factors-i.e., age, gender, smoking status, HDL-cholesterol concentrations, triglycerides levels, and HOMA-on the other, only age and IL-15 remained the predictors of IMT (β = 0.60, P = 0.0001 and β = 0.25, P = 0.024, respectively). There was no association of IL-15 with various anthropometric parameters nor with body fat distribution and severity of HS, also after adjusting for age. Age is resulted to be the main factor in the prediction of IMT and thus of early atherosclerosis. The prediction of IMT by IL-15 coupled with the lack of prediction by the well-known CAD risks is in agreement with recent data, which emphasizes the main role of the immune system in the onset/worsening of atherosclerosis, even though the role of visceral adiposity should be further deepened. Age and IL-15 levels were both predictors of early atherosclerosis in this population of obese patients with NAFLD, suggesting a possible role of this cytokine in the atherosclerosis process.
Collapse
Affiliation(s)
- Giovanni Tarantino
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, Naples, Italy
| | - Vincenzo Citro
- Department of General Medicine, “Umberto I” Hospital, Nocera Inferiore, Italy
| | - Clara Balsano
- Department of Clinical Medicine, Life, Health and Environmental Sciences-MESVA, University of L'Aquila, L'Aquila, Italy
| | | |
Collapse
|
78
|
Malhotra H, Garg V, Singh G. Biomarker Approach Towards Rheumatoid Arthritis Treatment. Curr Rheumatol Rev 2021; 17:162-175. [PMID: 33327920 DOI: 10.2174/1573397116666201216164013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 10/02/2020] [Accepted: 10/19/2020] [Indexed: 01/08/2023]
Abstract
Rheumatoid arthritis is an auto-immune disorder, recognized by cartilage as well as bone destruction, which causes irreversible joint deformities, which further results in functional limitations in the patient. Genes like HLA-DRB1 and PTPN22 are likely implicated in the genetic predisposition of rheumatoid arthritis pathology. The first and foremost clinical manifestation in a person with rheumatoid arthritis is joint destruction followed by cartilage and bone destruction caused by cell-cell interactions. The cell-cell interactions are thought to be initialized through the contact of antigen-presenting cells (APC) with CD4+ cells, leading to the progression of the disease. APC includes a complex of class ІІ major histocompatibility complex molecules along with peptide antigens and binds to the receptors present on the surface of T-cells. Further, the activation of macrophages is followed by the release of various pro-inflammatory cytokines such as IL-1 and TNF-α, which lead to the secretion of enzymes that degrade proteoglycan and collagen, which in turn, increase tissue degradation. Biomarkers like IL-6, IL-12, IL-8 and IL-18, 14-3-3η, RANKL, IFN-γ, IFN-β and TGF-β have been designated as key biomarkers in disease development and progression. The study of these biomarkers is very important as they act as a molecular indicator of pathological processes that aggravate the disease.
Collapse
Affiliation(s)
- Hitesh Malhotra
- Chandigarh College of Pharmacy Landran, Mohali, Punjab, India
| | - Vandana Garg
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
79
|
Abstract
The CD8+ T cell noncytotoxic antiviral response (CNAR) was discovered during studies of asymptomatic HIV-infected subjects more than 30 years ago. In contrast to CD8+ T cell cytotoxic lymphocyte (CTL) activity, CNAR suppresses HIV replication without target cell killing. This activity has characteristics of innate immunity: it acts on all retroviruses and thus is neither epitope specific nor HLA restricted. The HIV-associated CNAR does not affect other virus families. It is mediated, at least in part, by a CD8+ T cell antiviral factor (CAF) that blocks HIV transcription. A variety of assays used to measure CNAR/CAF and the effects on other retrovirus infections are described. Notably, CD8+ T cell noncytotoxic antiviral responses have now been observed with other virus families but are mediated by different cytokines. Characterizing the protein structure of CAF has been challenging despite many biologic, immunologic, and molecular studies. It represents a low-abundance protein that may be identified by future next-generation sequencing approaches. Since CNAR/CAF is a natural noncytotoxic activity, it could provide promising strategies for HIV/AIDS therapy, cure, and prevention.
Collapse
Affiliation(s)
- Maelig G Morvan
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Fernando C Teque
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | | | - Jay A Levy
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
80
|
Isvoranu G, Surcel M, Munteanu AN, Bratu OG, Ionita-Radu F, Neagu MT, Chiritoiu-Butnaru M. Therapeutic potential of interleukin-15 in cancer (Review). Exp Ther Med 2021; 22:675. [PMID: 33986840 PMCID: PMC8112152 DOI: 10.3892/etm.2021.10107] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 02/24/2021] [Indexed: 12/21/2022] Open
Abstract
The immune system is dysfunctional in cancer, and therapeutic approaches designated to restore immunity and increase long-term overall survival are desirable. The role of immunotherapy is to trigger the immune system to recognize and destroy tumor cells. Interleukin-15 (IL-15) is a member of the common gamma-chain (γc) cytokines that promote the differentiation and expansion of T cells, B cells and natural killer (NK) cells, leading to enhanced antitumor responses. This suggests that IL-15 is a promising candidate for anticancer therapy. Renewed interest in cancer immunotherapy has led to an increased number of preclinical studies and clinical trials that have investigated the reliability and potency of IL-15-based agents, not only as single therapy, but also in combination with others. This review provides a description of these studies which show the advantages and disadvantages of IL-15 as an immunotherapeutic agent. We present here the role of IL-15 and pharmacologically improved IL-15 superagonists as a single treatment or in combination with other therapeutic agents.
Collapse
Affiliation(s)
- Gheorghita Isvoranu
- Department of Animal Husbandry, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania
| | - Mihaela Surcel
- Department of Immunology, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania
| | - Adriana Narcisa Munteanu
- Department of Immunology, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania.,Department of Doctoral School of Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Ovidiu Gabriel Bratu
- Department of Clinical Department III, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Clinic of Urology, 'Dr. Carol Davila' University Emergency Central Military Hospital, 010825 Bucharest, Romania.,Academy of Romanian Scientists, 050094 Bucharest, Romania
| | - Florentina Ionita-Radu
- Department of Gastroenterology, 'Dr. Carol Davila' University Emergency Central Military Hospital, 010825 Bucharest, Romania
| | - Monica Teodora Neagu
- Department of Immunology, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania.,Department of Doctoral School of Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Marioara Chiritoiu-Butnaru
- Department of Molecular Cell Biology, Institute of Biochemistry of the Romanian Academy (IBAR), 060031 Bucharest, Romania
| |
Collapse
|
81
|
Zhao Y, Cai C, Samir J, Palgen JL, Keoshkerian E, Li H, Bull RA, Luciani F, An H, Lloyd AR. Human CD8 T-stem cell memory subsets phenotypic and functional characterization are defined by expression of CD122 or CXCR3. Eur J Immunol 2021; 51:1732-1747. [PMID: 33844287 DOI: 10.1002/eji.202049057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/07/2021] [Accepted: 04/07/2021] [Indexed: 11/12/2022]
Abstract
Long-lived T-memory stem cells (TSCM ) are key to both naturally occurring and vaccine-conferred protection against infection. These cells are characterized by the CD45RA+ CCR7+ CD95+ phenotype. Significant heterogeneity within the TSCM population is recognized, but distinguishing surface markers and functional characterization of potential subsets are lacking. Human CD8 TSCM subsets were identified in healthy subjects who had been previously exposed to CMV or Influenza (Flu) virus in flow cytometry by expression of CD122 or CXCR3, and then characterized in proliferation, multipotency, self-renewal, and intracellular cytokine production (TNF-α, IL-2, IFN-γ), together with transcriptomic profiles. The TSCM CD122hi -expressing subset (versus CD122lo ) demonstrated greater proliferation, greater multipotency, and enhanced polyfunctionality with higher frequencies of triple positive (TNF-α, IL-2, IFN-γ) cytokine-producing cells upon exposure to recall antigen. The TSCM CXCR3lo subpopulation also had increased proliferation and polyfunctional cytokine production. Transcriptomic analysis further showed that the TSCM CD122hi population had increased expression of activation and homing molecules, such as Ccr6, Cxcr6, Il12rb, and Il18rap, and downregulated cell proliferation inhibitors, S100A8 and S100A9. These data reveal that the TSCM CD122hi phenotype is associated with increased proliferation, enhanced multipotency and polyfunctionality with an activated memory-cell like transcriptional profile, and hence, may be favored for induction by immunization and for adoptive immunotherapy.
Collapse
Affiliation(s)
- Yanran Zhao
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Curtis Cai
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Jerome Samir
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Jean-Louis Palgen
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Elizabeth Keoshkerian
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Hui Li
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Rowena A Bull
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Fabio Luciani
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia.,School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Hongyan An
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Andrew R Lloyd
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| |
Collapse
|
82
|
Prasad M, Wojciech L, Brzostek J, Hu J, Chua YL, Tung DWH, Yap J, Rybakin V, Gascoigne NRJ. Expansion of an Unusual Virtual Memory CD8 + Subpopulation Bearing Vα3.2 TCR in Themis-Deficient Mice. Front Immunol 2021; 12:644483. [PMID: 33897691 PMCID: PMC8058184 DOI: 10.3389/fimmu.2021.644483] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/19/2021] [Indexed: 11/23/2022] Open
Abstract
Deletion of the gene for Themis affects T cell selection in the thymus, which would be expected to affect the TCR repertoire. We found an increased proportion of cells expressing Vα3.2 (TRAV9N-3) in the peripheral CD8+ T cell population in mice with germline Themis deficiency. Analysis of the TCRα repertoire indicated it was generally reduced in diversity in the absence of Themis, whereas the diversity of sequences using the TRAV9N-3 V-region element was increased. In wild type mice, Vα3.2+ cells showed higher CD5, CD6 and CD44 expression than non-Vα3-expressing cells, and this was more marked in cells from Themis-deficient mice. This suggested a virtual memory phenotype, as well as a stronger response to self-pMHC. The Vα3.2+ cells responded more strongly to IL-15, as well as showing bystander effector capability in a Listeria infection. Thus, the unusually large population of Vα3.2+ CD8+ T cells found in the periphery of Themis-deficient mice reflects not only altered thymic selection, but also allowed identification of a subset of bystander-competent cells that are also present in wild-type mice.
Collapse
Affiliation(s)
- Mukul Prasad
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lukasz Wojciech
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Joanna Brzostek
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| | - Jianfang Hu
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| | - Yen Leong Chua
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Desmond Wai Hon Tung
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jiawei Yap
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vasily Rybakin
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| | - Nicholas R. J. Gascoigne
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
83
|
Iwamoto N, Patel B, Song K, Mason R, Bolivar-Wagers S, Bergamaschi C, Pavlakis GN, Berger E, Roederer M. Evaluation of chimeric antigen receptor T cell therapy in non-human primates infected with SHIV or SIV. PLoS One 2021; 16:e0248973. [PMID: 33752225 PMCID: PMC7984852 DOI: 10.1371/journal.pone.0248973] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/08/2021] [Indexed: 01/06/2023] Open
Abstract
Achieving a functional cure is an important goal in the development of HIV therapy. Eliciting HIV-specific cellular immune responses has not been sufficient to achieve durable removal of HIV-infected cells due to the restriction on effective immune responses by mutation and establishment of latent reservoirs. Chimeric antigen receptor (CAR) T cells are an avenue to potentially develop more potent redirected cellular responses against infected T cells. We developed and tested a range of HIV- and SIV-specific chimeric antigen receptor (CAR) T cell reagents based on Env-binding proteins. In general, SHIV/SIV CAR T cells showed potent viral suppression in vitro, and adding additional CAR molecules in the same transduction resulted in more potent viral suppression than single CAR transduction. Importantly, the primary determinant of virus suppression potency by CAR was the accessibility to the Env epitope, and not the neutralization potency of the binding moiety. However, upon transduction of autologous T cells followed by infusion in vivo, none of these CAR T cells impacted either acquisition as a test of prevention, or viremia as a test of treatment. Our study illustrates limitations of the CAR T cells as possible antiviral therapeutics.
Collapse
Affiliation(s)
- Nami Iwamoto
- ImmunoTechnology Section, Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Bhavik Patel
- ImmunoTechnology Section, Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Kaimei Song
- ImmunoTechnology Section, Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rosemarie Mason
- ImmunoTechnology Section, Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sara Bolivar-Wagers
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Cristina Bergamaschi
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Edward Berger
- Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mario Roederer
- ImmunoTechnology Section, Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
84
|
Wang X, Zhao XY. Transcription Factors Associated With IL-15 Cytokine Signaling During NK Cell Development. Front Immunol 2021; 12:610789. [PMID: 33815365 PMCID: PMC8013977 DOI: 10.3389/fimmu.2021.610789] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 03/01/2021] [Indexed: 12/16/2022] Open
Abstract
Natural killer (NK) cells are lymphocytes primarily involved in innate immunity and possess important functional properties in anti-viral and anti-tumor responses; thus, these cells have broad potential for clinical utilization. NK cells originate from hematopoietic stem cells (HSCs) through the following two independent and continuous processes: early commitment from HSCs to IL-15-responsive NK cell progenitors (NKPs) and subsequent differentiation into mature NK cells in response to IL-15. IL-15 is the most important cytokine for NK cell development, is produced by both hematopoietic and nonhematopoietic cells, and functions through a distinct delivery process termed transpresentation. Upon being transpresented to NK cells, IL-15 contributes to NK cell development via the activation of several downstream signaling pathways, including the Ras-MEK-MAPK, JAK-STAT5, and PI3K-ATK-mTOR pathways. Nonetheless, the exact role of IL-15 in NK cell development has not been discussed in a consecutive and comprehensive manner. Here, we review current knowledge about the indispensable role of IL-15 in NK cell development and address which cells produce IL-15 to support NK cell development and when IL-15 exerts its function during multiple developmental stages. Specifically, we highlight how IL-15 supports NK cell development by elucidating the distinct transpresentation of IL-15 to NK cells and revealing the downstream target of IL-15 signaling during NK cell development.
Collapse
Affiliation(s)
- Xiang Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Engineering Laboratory for Cellular Therapy, Beijing, China
| |
Collapse
|
85
|
Heterodimeric IL-15 in Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13040837. [PMID: 33671252 PMCID: PMC7922495 DOI: 10.3390/cancers13040837] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The rapidly expanding field of cancer immunotherapy uses diverse technologies, including cytokines, T cells, and antibody administration, with the aim to induce effective immune responses leading to tumor control. Interleukin-15 (IL-15), a cytokine discovered in 1994, supports the homeostasis of cytotoxic immune cells and shows promise as an anti-tumor agent. Many studies have elucidated IL-15 synthesis, regulation and biological function and explored its therapeutic efficacy in preclinical cancer models. Escherichia coli-derived single-chain IL-15 was tested in the first in-human trial in cancer patients. Its effects were limited by the biology of IL-15, which in vivo comprises a complex of the IL-15 chain with the IL-15 receptor alpha (IL-15Rα) chain, together forming the IL-15 heterodimer (hetIL-15). Currently, single-chain IL-15 and several heterodimeric IL-15:IL-15Rα variants (hetIL-15, N-803 and RLI) are being tested in clinical trials. This review presents a summary of contemporary preclinical and clinical research on IL-15. Abstract Immunotherapy has emerged as a valuable strategy for the treatment of many cancer types. Interleukin-15 (IL-15) promotes the growth and function of cytotoxic CD8+ T and natural killer (NK) cells. It also enhances leukocyte trafficking and stimulates tumor-infiltrating lymphocytes expansion and activity. Bioactive IL-15 is produced in the body as a heterodimeric cytokine, comprising the IL-15 and the so-called IL-15 receptor alpha chain that are together termed “heterodimeric IL-15” (hetIL-15). hetIL-15, closely resembling the natural form of the cytokine produced in vivo, and IL-15:IL-15Rα complex variants, such as hetIL-15Fc, N-803 and RLI, are the currently available IL-15 agents. These molecules have showed favorable pharmacokinetics and biological function in vivo in comparison to single-chain recombinant IL-15. Preclinical animal studies have supported their anti-tumor activity, suggesting IL-15 as a general method to convert “cold” tumors into “hot”, by promoting tumor lymphocyte infiltration. In clinical trials, IL-15-based therapies are overall well-tolerated and result in the expansion and activation of NK and memory CD8+ T cells. Combinations with other immunotherapies are being investigated to improve the anti-tumor efficacy of IL-15 agents in the clinic.
Collapse
|
86
|
Guo S, Smeltz RB, Nanajian A, Heller R. IL-15/IL-15Rα Heterodimeric Complex as Cancer Immunotherapy in Murine Breast Cancer Models. Front Immunol 2021; 11:614667. [PMID: 33628206 PMCID: PMC7897681 DOI: 10.3389/fimmu.2020.614667] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/15/2020] [Indexed: 01/22/2023] Open
Abstract
Interleukin 15 (IL-15) has been evaluated as a potential treatment for solid tumors in clinical trials, but the effectiveness of systemic IL-15 administration as a monotherapy has not been realized. IL-15 receptor alpha (IL-15Rα) can stabilize IL-15 and enhance its bioactivity. The goal of this study was to examine the activity of IL-15/IL-15Rα complex (IL-15cx) to CD8+ T cells and evaluate its potential efficacy in murine breast cancer models. The antitumor efficacy was studied in mouse mammary carcinoma models (Her2/neu transgenic and 4T1-luc mammary cancers) treated with systemic recombinant protein with/without the depletion of myeloid-derived suppressor cells or intra-tumoral gene electrotransfer (GET). IL-15cx shows superior in vivo bioactivity to expand CD8 T cells in comparison to an equimolar single chain IL-15. T-bet is partially involved in CD8 T cell expansion ex vivo and in vivo due to IL-15 or IL-15cx. Intraperitoneal administration of IL-15cx results in a moderate inhibition of breast cancer growth that is associated with an increase in the frequency of cytotoxic CD8 T cells and the improvement of their function. The depletion of myeloid-derived suppressor cells (MDSCs) has no impact on mouse breast cancer growth. IL-15cx treatment diminishes MDSCs in murine tumors. However, it also antagonizes the effects of anti-Gr-1 depleting antibodies. Intratumoral GET with plasmid IL-15/IL-15Rα leads to a long-term survival benefit in 4T1 mammary carcinoma model. An early increase of local cytotoxic cells correlates with GET treatment and an increase of long-term memory T cells results from animals with complete tumor regression. Systemic and local administration of IL-15cx shows two distinct therapeutic responses, a moderate tumor growth inhibition or heterogeneous tumor regressions with survival improvement. Further studies are warranted to improve the efficacy of IL-15cx as an immunotherapy for breast cancer.
Collapse
Affiliation(s)
- Siqi Guo
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States.,Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, VA, United States
| | - Ronald B Smeltz
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, VA, United States
| | - Anthony Nanajian
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States
| | - Richard Heller
- Department of Medical Engineering, University of South Florida, Tampa, FL, United States
| |
Collapse
|
87
|
Abstract
Clinical trials have demonstrated that an increased number of effector cells, especially tumor-specific T cells, is positively linked with patients’ prognosis. Although the discovery of checkpoint inhibitors (CPIs) has led to encouraging progress in cancer immunotherapy, the lack of either T cells or targets for CPIs is a limitation for patients with poor prognosis. Since interleukin (IL)-2 and IL-7 are cytokines that target many aspects of T-cell responses, they have been used to treat cancers. In this review, we focus on the basic biology of how these cytokines regulate T-cell response and on the clinical trials using the cytokines against cancer. Further, we introduce several recent studies that aim to improve cytokines’ biological activities and find the strategy for combination with other therapeutics.
Collapse
Affiliation(s)
- Ji-Hae Kim
- Department of Life Sciences, Pohang University of Science and Technology(POSTECH), Pohang 37673, Korea
| | - Kun-Joo Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology(POSTECH), Pohang 37673, Korea
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology(POSTECH), Pohang 37673, Korea
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology(POSTECH), Pohang 37673, Korea
| |
Collapse
|
88
|
Chen Z, Yang Y, Neo SY, Shi H, Chen Y, Wagner AK, Larsson K, Tong L, Jakobsson PJ, Alici E, Wu J, Cao Y, Wang K, Liu LL, Mao Y, Sarhan D, Lundqvist A. Phosphodiesterase 4A confers resistance to PGE2-mediated suppression in CD25 + /CD54 + NK cells. EMBO Rep 2021; 22:e51329. [PMID: 33480074 PMCID: PMC7926252 DOI: 10.15252/embr.202051329] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 12/22/2022] Open
Abstract
Inadequate persistence of tumor‐infiltrating natural killer (NK) cells is associated with poor prognosis in cancer patients. The solid tumor microenvironment is characterized by the presence of immunosuppressive factors, including prostaglandin E2 (PGE2), that limit NK cell persistence. Here, we investigate if the modulation of the cytokine environment in lung cancer with IL‐2 or IL‐15 renders NK cells resistant to suppression by PGE2. Analyzing Cancer Genome Atlas (TCGA) data, we found that high NK cell gene signatures correlate with significantly improved overall survival in patients with high levels of the prostaglandin E synthase (PTGES). In vitro, IL‐15, in contrast to IL‐2, enriches for CD25+/CD54+ NK cells with superior mTOR activity and increased expression of the cAMP hydrolyzing enzyme phosphodiesterase 4A (PDE4A). Consequently, this distinct population of NK cells maintains their function in the presence of PGE2 and shows an increased ability to infiltrate lung adenocarcinoma tumors in vitro and in vivo. Thus, strategies to enrich CD25+/CD54+ NK cells for adoptive cell therapy should be considered.
Collapse
Affiliation(s)
- Ziqing Chen
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ying Yang
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China.,School of Medicine, Zhejiang University, Hangzhou, China
| | - Shi Y Neo
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Hao Shi
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Yi Chen
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Arnika K Wagner
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Karin Larsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Le Tong
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Evren Alici
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Jing Wu
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Kai Wang
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Lisa L Liu
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Yumeng Mao
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Dhifaf Sarhan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
89
|
Suzuki T, Kato Y, Kobayashi A, Suzuki K. Protective effects of aqueous extracts of Rhizopus oryzae on atopic dermatitis in NC/Nga mice. FOOD SCIENCE AND TECHNOLOGY RESEARCH 2021. [DOI: 10.3136/fstr.27.725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
| | - Yukiko Kato
- School of Veterinary Medicine, Azabu University
| | | | | |
Collapse
|
90
|
Du Y, Fang Q, Zheng SG. Regulatory T Cells: Concept, Classification, Phenotype, and Biological Characteristics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1278:1-31. [PMID: 33523440 DOI: 10.1007/978-981-15-6407-9_1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Regulatory T cells (Treg) play an indispensable role in maintaining the body's immune nonresponse to self-antigens and suppressing the body's unwarranted and potentially harmful immune responses. Their absence, reduction, dysfunction, transformation, and instability can lead to numerous autoimmune diseases. There are several distinct subtypes of the Treg cells, although they share certain biological characteristics and have unique phenotypes with different regulatory functions, as well as mechanistic abilities. In this book chapter, we introduce the latest advances in Treg cell subtypes pertaining to classification, phenotype, biological characteristics, and mechanisms. We also highlight the relationship between Treg cells and various diseases, including autoimmune, infectious, as well as tumors and organ transplants.
Collapse
Affiliation(s)
- Yang Du
- Department of Pathology and Physiopathology, Guilin Medical University, Guilin, Guangxi, China.,Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Qiannan Fang
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Song-Guo Zheng
- Department of Internal Medicine, Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
91
|
Yang Y, Lundqvist A. Immunomodulatory Effects of IL-2 and IL-15; Implications for Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12123586. [PMID: 33266177 PMCID: PMC7761238 DOI: 10.3390/cancers12123586] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
The type I cytokine family members interleukin-2 (IL-2) and IL-15 play important roles in the homeostasis of innate and adaptive immunity. Although IL-2 and IL-15 receptor complexes activate similar signal transduction cascades, triggering of these receptors results in different functional activities in lymphocytes. While IL-2 expands regulatory T cells and CD4+ helper T cells, IL-15 supports the development of central memory T cells and NK cells. Recent data have provided evidence that IL-2 and IL-15 differ in their ability to activate T and NK cells to resist various forms of immune suppression. The diverse roles of these two cytokines have on immune cells lead to critical therapeutic implications for cancer treatment. In this review, we discuss the distinct roles of IL-2 and IL-15 in activating various functions in T and NK cells with a particular focus on the signals that participate in the resistance of tumor-derived immune suppressive factors. Furthermore, we summarize current clinical applications of IL-2 and IL-15 in metastatic malignancies, either as monotherapy or in combination with other agents, and highlight the future trends for research on these cytokine-based immunotherapies.
Collapse
Affiliation(s)
- Ying Yang
- Department of Respiratory, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 310009, China;
- Department of Oncology-Pathology, Karolinska Institutet, S-17164 Stockholm, Sweden
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, S-17164 Stockholm, Sweden
- Correspondence:
| |
Collapse
|
92
|
Thiele D, La Gruta NL, Nguyen A, Hussain T. Hiding in Plain Sight: Virtually Unrecognizable Memory Phenotype CD8 + T cells. Int J Mol Sci 2020; 21:ijms21228626. [PMID: 33207648 PMCID: PMC7698292 DOI: 10.3390/ijms21228626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023] Open
Abstract
Virtual memory T (TVM) cells are a recently described population of conventional CD8+ T cells that, in spite of their antigen inexperience, express markers of T cell activation. TVM cells exhibit rapid responsiveness to both antigen-specific and innate stimuli in youth but acquire intrinsic antigen-specific response defects in the elderly. In this article, we review how the identification of TVM cells necessitates a re-evaluation of accepted paradigms for conventional memory T (TMEM) cells, the potential for heterogeneity within the TVM population, and the defining characteristics of TVM cells. Further, we highlight recent literature documenting the development of TVM cells as a distinct CD8+ T cell lineage as well their biological significance in the context of disease.
Collapse
|
93
|
Parga-Vidal L, van Gisbergen KPJM. Area under Immunosurveillance: Dedicated Roles of Memory CD8 T-Cell Subsets. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037796. [PMID: 32839203 DOI: 10.1101/cshperspect.a037796] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Immunological memory, defined as the ability to respond in an enhanced manner upon secondary encounter with the same pathogen, can provide substantial protection against infectious disease. The improved protection is mediated in part by different populations of memory CD8 T cells that are retained after primary infection. Memory cells persist in the absence of pathogen-derived antigens and enable secondary CD8 T-cell responses with accelerated kinetics and of larger magnitude after reencounter with the same pathogen. At least three subsets of memory T cells have been defined that are referred to as central memory CD8 T cells (Tcm), effector memory CD8 T cells (Tem), and tissue-resident memory CD8 T cells (Trm). Tcm and Tem are circulating memory T cells that mediate bodywide immune surveillance in search of invading pathogens. In contrast, Trm permanently reside in peripheral barrier tissues, where they form a stationary defensive line of sentinels that alert the immune system upon pathogen reencounter. The characterization of these different subsets has been instrumental in our understanding of the strategies that memory T cells employ to counter invading pathogens. It is clear that memory T cells not only have a numerical advantage over naive T cells resulting in improved protection in secondary responses, but also acquire distinct sets of competencies that assist in pathogen clearance. Nevertheless, inherent challenges are associated with the allocation of memory T cells to a limited number of subsets. The classification of memory T cells into Tcm, Tem, and Trm may not take into account the full extent of the heterogeneity that is observed in the memory population. Therefore, in this review, we will revisit the current classification of memory subsets, elaborate on functional and migratory properties attributed to Tcm, Tem, and Trm, and discuss how potential heterogeneity within these populations arises and persists.
Collapse
Affiliation(s)
- Loreto Parga-Vidal
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066CX Amsterdam, The Netherlands
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066CX Amsterdam, The Netherlands
| |
Collapse
|
94
|
Wilz SW. A clinical trial of IL-15 and IL-21 combination therapy for COVID-19 is warranted. Cytokine Growth Factor Rev 2020; 58:49-50. [PMID: 33199178 PMCID: PMC7583616 DOI: 10.1016/j.cytogfr.2020.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/20/2020] [Indexed: 01/18/2023]
Abstract
Previous studies of SARS-CoV-2 viral infection suggest that both the humoral and cytotoxic arms of the immune system are weak in patients with severe COVID-19 disease when compared to mild disease. A cytokine storm is also induced in severe disease. IL-15 has been shown to support the cytotoxic arm of the immune response. IL-21 has been shown to support both the cytotoxic and humoral arms of the immune response. In addition, in some settings, Il-21 has been shown to actually decrease IL-6 and TNF-alpha production, reducing the inflammatory proteins involved in the cytokine storm. Furthermore, in other settings, the combination of IL-15 and IL-21 has been shown to be more effective than either interleukin alone in promoting an effective immune response. Therefore, a clinical trial that examines the use of the combination of IL-15 and IL-21 for COVID-19 patients is warranted.
Collapse
Affiliation(s)
- Stephen W Wilz
- Boston VA Healthcare System, Pathology and Laboratory Medicine, 1400 VFW Parkway, West Roxbury, MA, 02132, United States.
| |
Collapse
|
95
|
Mikkilineni L, Kochenderfer JN. CAR T cell therapies for patients with multiple myeloma. Nat Rev Clin Oncol 2020; 18:71-84. [PMID: 32978608 DOI: 10.1038/s41571-020-0427-6] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2020] [Indexed: 12/21/2022]
Abstract
Despite several therapeutic advances over the past decade, multiple myeloma (MM) remains largely incurable, indicating a need for new treatment approaches. Chimeric antigen receptor (CAR) T cell therapy works by mechanisms distinct from those of other MM therapies and involves the modification of patient or donor T cells to target specific cell-surface antigens. B cell maturation antigen (BCMA) is expressed only on plasma cells, a small subset of B cells and MM cells, which makes it a suitable target antigen for such therapies. At the time of writing, data from >20 clinical trials involving anti-BCMA CAR T cells have demonstrated that patients with relapsed and/or refractory MM can achieve objective responses. These early investigations have been instrumental in demonstrating short-term safety and efficacy; however, most patients do not have disease remission lasting >18 months. Attempts to reduce or delay the onset of relapsed disease are underway and include identifying additional CAR T cell target antigens and methods of enhancing BCMA expression on MM cells. Engineering CAR T cells to enhance both the activity and safety of treatment continues to be a promising avenue for improvement. In this Review we summarize data from clinical trials that have been carried out to date, describe novel antigens that could be targeted in the future, and highlight potential future innovations that could enhance the efficacy and/or reduce the toxicities associated with CAR T cell therapies.
Collapse
Affiliation(s)
- Lekha Mikkilineni
- Surgery Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA.
| | | |
Collapse
|
96
|
Ataca Atilla P, McKenna MK, Tashiro H, Srinivasan M, Mo F, Watanabe N, Simons BW, McLean Stevens A, Redell MS, Heslop HE, Mamonkin M, Brenner MK, Atilla E. Modulating TNFα activity allows transgenic IL15-Expressing CLL-1 CAR T cells to safely eliminate acute myeloid leukemia. J Immunother Cancer 2020; 8:jitc-2020-001229. [PMID: 32938629 PMCID: PMC7497527 DOI: 10.1136/jitc-2020-001229] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2020] [Indexed: 12/12/2022] Open
Abstract
Background C-type lectin-like molecule 1 (CLL-1) is highly expressed in acute myeloid leukemia (AML) but is absent in primitive hematopoietic progenitors, making it an attractive target for a chimeric antigen receptor (CAR) T-cell therapy. Here, we optimized our CLL-1 CAR for anti-leukemic activity in mouse xenograft models of aggressive AML. Methods First, we optimized the CLL-1 CAR using different spacer, transmembrane and costimulatory sequences. We used a second retroviral vector to coexpress transgenic IL15. We measured the effects of each construct on T cell phenotype and sequential (recursive) co culture assays with tumor cell targets to determine the durability of the anti tumor activity by flow cytometry. We administered CAR T cells to mice engrafted with patient derived xenografts (PDX) and AML cell line and determined anti tumor activity by bioluminescence imaging and weekly bleeding, measured serum cytokines by multiplex analysis. After euthanasia, we examined formalin-fixed/paraffin embedded sections. Unpaired two-tailed Student’s t-tests were used and values of p<0.05 were considered significant. Survival was calculated using Mantel-Cox log-rank test. Results In vitro, CLL-1 CAR T cells with interleukin-15 (IL15) were less terminally differentiated (p<0.0001) and had superior expansion compared with CD28z-CD8 CAR T cells without IL15 (p<0.001). In both AML PDX and AML cell line animal models, CLL-1 CAR T coexpressing transgenic IL15 initially expanded better than CD28z-CD8 CAR T without IL15 (p<0.0001), but produced severe acute toxicity associated with high level production of human tumor necrosis factor α (TNFα), IL15 and IL2. Histopathology showed marked inflammatory changes with tissue damage in lung and liver. This acute toxicity could be managed by two strategies, individually or in combination. The excessive TNF alpha secretion could be blocked with anti-TNF alpha antibody, while excessive T cell expansion could be arrested by activation of an inducible caspase nine safety switch by administration of dimerizing drug. Both strategies successfully prolonged tumor-free survival. Conclusion Combinatorial treatment with a TNFα blocking antibody and subsequent activation of the caspase-9 control switch increased the expansion, survival and antileukemic potency of CLL-1 CAR T-cells expressing transgenic IL15 while avoiding the toxicities associated with excessive cytokine production and long-term accumulation of activated T-cells.
Collapse
Affiliation(s)
- Pinar Ataca Atilla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Mary K McKenna
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Haruko Tashiro
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | | | - Feiyan Mo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Norihiro Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Brian Wesley Simons
- Center for Comparative Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Alexandra McLean Stevens
- Division of Pediatric Hematology/Oncology, Texas Children's Hospital, Houston, Texas, USA.,Division of Pediatric Hematology/Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Michele S Redell
- Division of Pediatric Hematology/Oncology, Texas Children's Hospital, Houston, Texas, USA.,Division of Pediatric Hematology/Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA.,Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Maksim Mamonkin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA.,Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Erden Atilla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
97
|
Cooke RE, Quinn KM, Quach H, Harrison S, Prince HM, Koldej R, Ritchie D. Conventional Treatment for Multiple Myeloma Drives Premature Aging Phenotypes and Metabolic Dysfunction in T Cells. Front Immunol 2020; 11:2153. [PMID: 33013907 PMCID: PMC7494758 DOI: 10.3389/fimmu.2020.02153] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/07/2020] [Indexed: 12/17/2022] Open
Abstract
New diagnoses of multiple myeloma (MM) tend to occur after the age of 60, by which time thymic output is severely reduced. As a consequence, lymphocyte recovery after lymphopenia-inducing anti-MM therapies relies on homeostatic proliferation of peripheral T cells rather than replenishment by new thymic emigrants. To assess lymphocyte recovery and phenotype in patients with newly diagnosed MM (NDMM) and relapsed/refractory MM (RRMM), we tracked CD4+ and CD8+ T cell populations at serial time points throughout treatment and compared them to age-matched healthy donors (HD). Anti-MM therapies and autologous stem cell transplant (ASCT) caused a permanent reduction in the CD4:8 ratio, a decrease in naïve CD4+ T cells, and an increase in effector memory T cells and PD1-expressing CD4+ T cells. Transcriptional profiling highlighted that genes associated with fatty acid β-oxidation were upregulated in T cells in RRMM, suggesting increased reliance on mitochondrial respiration. High mitochondrial mass was seen in all T cell subsets in RRMM but with relatively suppressed reactive oxygen species and mitochondrial membrane potential, indicating mitochondrial dysfunction. These findings highlight that anti-MM and ASCT therapies perturb the composition of the T cell compartment and drive substantial metabolic remodeling, which may affect the fitness of T cells for immunotherapies. This is particularly pertinent to chimeric antigen receptor (CAR)-T therapy, which might be more efficacious if T cells were stored prior to ASCT rather than at relapse.
Collapse
Affiliation(s)
- Rachel Elizabeth Cooke
- Australian Cancer Research Foundation Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Kylie Margaret Quinn
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Hang Quach
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Simon Harrison
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Henry Miles Prince
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Rachel Koldej
- Australian Cancer Research Foundation Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - David Ritchie
- Australian Cancer Research Foundation Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
98
|
Fara A, Mitrev Z, Rosalia RA, Assas BM. Cytokine storm and COVID-19: a chronicle of pro-inflammatory cytokines. Open Biol 2020; 10:200160. [PMID: 32961074 PMCID: PMC7536084 DOI: 10.1098/rsob.200160] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) has swept the world, unlike any other pandemic in the last 50 years. Our understanding of the disease has evolved rapidly since the outbreak; disease prognosis is influenced mainly by multi-organ involvement. Acute respiratory distress syndrome, heart failure, renal failure, liver damage, shock and multi-organ failure are strongly associated with morbidity and mortality. The COVID-19 disease pathology is plausibly linked to the hyperinflammatory response of the body characterized by pathological cytokine levels. The term 'cytokine storm syndrome' is perhaps one of the critical hallmarks of COVID-19 disease severity. In this review, we highlight prominent cytokine families and their potential role in COVID-19, the type I and II interferons, tumour necrosis factor and members of the Interleukin family. We address various changes in cellular components of the immune response corroborating with changes in cytokine levels while discussing cytokine sources and biological functions. Finally, we discuss in brief potential therapies attempting to modulate the cytokine storm.
Collapse
Affiliation(s)
| | - Zan Mitrev
- Department of Clinical Research, Zan Mitrev Clinic, St. Bledski Dogovor 8, 1000 Skopje, The Republic of North Macedonia
| | - Rodney Alexander Rosalia
- Department of Clinical Research, Zan Mitrev Clinic, St. Bledski Dogovor 8, 1000 Skopje, The Republic of North Macedonia
| | - Bakri M. Assas
- Faculty of Applied Medical Sciences, Department of Medical Laboratory Technology, Immunology group, King Abdul Aziz University, Jeddah, Saudi Arabia
| |
Collapse
|
99
|
Chen Y, Yan Y, Liu H, Qiu F, Liang CL, Zhang Q, Huang RY, Han L, Lu C, Dai Z. Dihydroartemisinin ameliorates psoriatic skin inflammation and its relapse by diminishing CD8 + T-cell memory in wild-type and humanized mice. Theranostics 2020; 10:10466-10482. [PMID: 32929360 PMCID: PMC7482806 DOI: 10.7150/thno.45211] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022] Open
Abstract
Conventional immunosuppressants cause side effects and do not prevent the recurrence of autoimmune diseases. Moreover, they may not inhibit autoimmunity mediated by pathogenic memory T-cells. Dihydroartemisinin (DHA) has been shown to regulate autoimmunity. However, it remains unknown whether DHA impacts psoriasis and its recurrence. The objective of this study was to determine therapeutic effects of DHA on psoriasis and its relapse as well as its underlying mechanisms. Methods: We established animal models of imiquimod (IMQ)-induced psoriasis-like wild-type mice and humanized NSG mice receiving lesional human skin from patients with psoriasis. Many immunoassays, including immunohistochemistry, flow cytometry, quantitative RT-PCR and Western blotting, were performed. Results: We found that DHA not only ameliorated acute skin lesion of psoriatic mice, but also alleviated its recurrence by diminishing CD8+ central memory T (TCM) and CD8+ resident memory T (TRM) cells. It attenuated epidermal pathology and T-cell infiltration in the skin of IMQ-induced psoriatic mice while suppressing expression of IL-15, IL-17 and other proinflammatory cytokines in the skin. Surprisingly, DHA reduced the frequency and number of CD8+, but not CD4+, subset of CD44highCD62Lhigh TCM in psoriatic mice, whereas methotrexate (MTX) lowered CD4+, but not CD8+, TCM frequency and number. Indeed, DHA, but not MTX, downregulated eomesodermin (EOMES) and BCL-6 expression in CD8+ T-cells. Furthermore, DHA, but not MTX, reduced the presence of CD8+CLA+, CD8+CD69+ or CD8+CD103+ TRM cells in mouse skin. Interestingly, treatment with DHA, but not MTX, during the first onset of psoriasis largely prevented psoriasis relapse induced by low doses of IMQ two weeks later. Administration of recombinant IL-15 or CD8+, but not CD4+, TCM cells resulted in complete recurrence of psoriasis in mice previously treated with DHA. Finally, we demonstrated that DHA alleviated psoriatic human skin lesions in humanized NSG mice grafted with lesional skin from psoriatic patients while reducing human CD8+ TCM and CD103+ TRM cells in humanized mice. Conclusion: We have provided the first evidence that DHA is advantageous over MTX in preventing psoriasis relapse by reducing memory CD8+ T-cells.
Collapse
Affiliation(s)
- Yuchao Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Yuhong Yan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Huazhen Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Feifei Qiu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Chun-Ling Liang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Qunfang Zhang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Run-Yue Huang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Ling Han
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Chuanjian Lu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| | - Zhenhua Dai
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong 510006, China
| |
Collapse
|
100
|
Barut GT, Lischer HEL, Bruggmann R, Summerfield A, Talker SC. Transcriptomic profiling of bovine blood dendritic cells and monocytes following TLR stimulation. Eur J Immunol 2020; 50:1691-1711. [PMID: 32592404 DOI: 10.1002/eji.202048643] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/11/2020] [Accepted: 06/26/2020] [Indexed: 11/06/2022]
Abstract
Dendritic cells (DC) and monocytes are vital for the initiation of innate and adaptive immune responses. Recently, we identified bona fide DC subsets in blood of cattle, revealing subset- and species-specific transcription of toll-like receptors (TLR). In the present study, we analyzed phenotypic and transcriptional responses of bovine DC subsets and monocytes to in vitro stimulation with four to six different TLR ligands. Bovine DC subsets, especially plasmacytoid DC (pDC), showed a clear increase of CCR7, CD25, CD40, CD80, CD86, and MHC-II expression both on mRNA and protein level. Flow cytometric detection of p38 MAPK phosphorylation 15 min after stimulation confirmed activation of DC subsets and monocytes in accordance with TLR gene expression. Whole-transcriptome sequencing of sorted and TLR-stimulated subsets revealed potential ligand- and subset-specific regulation of genes associated with inflammation, T-cell co-stimulation, migration, metabolic reprogramming, and antiviral activity. Gardiquimod was found to evoke strong responses both in DC subsets and monocytes, while Poly(I:C) and CpG preferentially triggered responses in cDC1 and pDC, respectively. This in-depth analysis of ligand responsiveness is essential for the rational design of vaccine adjuvants in cattle, and provides a solid basis for comparative studies on DC and monocyte biology across species.
Collapse
Affiliation(s)
- G Tuba Barut
- Institute of Virology and Immunology, Bern & Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Heidi E L Lischer
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Artur Summerfield
- Institute of Virology and Immunology, Bern & Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Stephanie C Talker
- Institute of Virology and Immunology, Bern & Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|