1
|
SantaCruz-Calvo S, Saraswat S, Kalantar GH, Zukowski E, Marszalkowski H, Javidan A, Gholamrezaeinejad F, Bharath LP, Kern PA, Zhang XD, Nikolajczyk BS. A unique inflammaging profile generated by T cells from people with obesity is metformin resistant. GeroScience 2024:10.1007/s11357-024-01441-4. [PMID: 39708215 DOI: 10.1007/s11357-024-01441-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 11/07/2024] [Indexed: 12/23/2024] Open
Abstract
The alarmingly high prevalence of obesity in older adults coupled with the negative health effects of chronic inflammation in both obesity and aging highlight the importance of studies investigating the impacts of obesity on age-related inflammation. Since shifts in peripheral T-cell metabolism and function drive systemic inflammation in both obesity and aging, we hypothesize that obesity impacts the Th17-dominated inflammaging profile we identified in lean subjects and thus modifies the anti-inflammatory effects of geroprotective drugs like metformin. New cytokine profiling data showed that CD4+ T cells from older people with obesity generate a profile that specifically excludes Th17 cytokines. Metformin failed to change the age-associated T-cell profile in obesity, despite lowering both mitochondrial respiration and reactive oxygen species (ROS) production. Metformin did not improve macroautophagy in T cells from older people with obesity, in sharp contrast to the ability of metformin to promote autophagy in T cells from older lean subjects. These data indicate that body mass index modifies the mechanisms supporting inflammaging in T cells from older subjects, and that metformin-mediated restoration of redox balance is insufficient to stem obesity-associated inflammaging. We conclude that obesity fundamentally changes the mechanisms that promote inflammaging, and thus obesity becomes a critical consideration for clinical trials of geroprotective agents such as metformin.
Collapse
Affiliation(s)
- S SantaCruz-Calvo
- Department of Pharmacology & Nutritional Sciences, Diabetes and Obesity Research Priority Area, University of Kentucky, Lexington, KY, USA
| | - S Saraswat
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - G H Kalantar
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - E Zukowski
- Department of Nutrition & Public Health, Merrimack College, North Andover, MA, USA
| | - H Marszalkowski
- Department of Biology, Merrimack College, North Andover, MA, USA
| | - A Javidan
- Department of Pharmacology & Nutritional Sciences, Diabetes and Obesity Research Priority Area, University of Kentucky, Lexington, KY, USA
| | - F Gholamrezaeinejad
- Department of Pharmacology & Nutritional Sciences, Diabetes and Obesity Research Priority Area, University of Kentucky, Lexington, KY, USA
| | - L P Bharath
- Department of Nutrition & Public Health, Merrimack College, North Andover, MA, USA
| | - P A Kern
- Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, KY, USA
| | - X D Zhang
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, KY, USA
| | - B S Nikolajczyk
- Department of Pharmacology & Nutritional Sciences, Diabetes and Obesity Research Priority Area, University of Kentucky, Lexington, KY, USA.
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, KY, USA.
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
2
|
Nejabat S, Khomartash MS, Mohammadimehr M, Adloo Z, Zanchi FB, Ghorbani M, Nezafat N. Immunoinformatics approach: Developing a multi-epitope vaccine with novel carriers targeting monkeypox virus. FASEB J 2024; 38:e70257. [PMID: 39679938 DOI: 10.1096/fj.202400757rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/12/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
Since May 2022, the global spread of monkeypox virus (MPXV) has presented a significant threat to public health. Despite this, there are limited preventive measures available. In this study, different computational tools were employed to design a multi-epitope vaccine targeting MPXV. Three key MPXV proteins, M1R, B6R, and F3L, were chosen for epitope selection, guided by bioinformatic analyses to identify immunodominant epitopes for T- and B-cell activation. To enhance immune stimulation and facilitate targeted delivery of the vaccine to specific cells, the selected epitopes were linked to novel carriers, including the extracellular domain of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), a 12-mer Clec9a binding peptide (CBP-12), and a Toll-like receptor 2 (TLR2) peptide ligand. The designed vaccine construct exhibited strong antigenicity along with nonallergenic and nontoxic properties, with favorable physicochemical characteristics. The validated vaccine's tertiary structure underwent evaluation for interactions with CD80/86, Clec9a, and TLR2 through molecular docking and molecular dynamics simulation. The results ensured the vaccine's stability and high affinity for the aforementioned receptors. In silico immune simulations studies revealed robust innate and adaptive immune responses, including enhanced mucosal immunity essential for protection against MPXV. Ultimately, the DNA sequence of the vaccine construct was synthesized and successfully cloned into the pET-22b(+) vector. Our study, through integration of computational predictions, suggests the proposed vaccine's potential efficacy in safeguarding against MPXV; however, further in vitro and in vivo validations are imperative to assess real-world effectiveness and safety.
Collapse
Affiliation(s)
- Sajjad Nejabat
- Science and Technology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mojgan Mohammadimehr
- Infectious Diseases Research Center, AJA University of Medical Sciences, Tehran, Iran
- Department of Laboratory Sciences, Faculty of Paramedicine, AJA University of Medical Sciences, Tehran, Iran
| | - Zahra Adloo
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fernando Berton Zanchi
- Laboratório de Bioinformática e Química Medicinal (LABIOQUIM), Fundação Oswaldo Cruz Rondônia, Porto Velho, Brazil
| | - Mahdi Ghorbani
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, AJA University of Medical Sciences, Tehran, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
3
|
Tao Z, Chyra Z, Kotulová J, Celichowski P, Mihályová J, Charvátová S, Hájek R. Impact of T cell characteristics on CAR-T cell therapy in hematological malignancies. Blood Cancer J 2024; 14:213. [PMID: 39627220 PMCID: PMC11615218 DOI: 10.1038/s41408-024-01193-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/06/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment paradigms for hematological malignancies. However, more than half of these patients cannot achieve sustainable tumor control, partially due to the inadequate potency of CAR-T cells in eradicating tumor cells. T cells are crucial components of the anti-tumor immune response, and multiple intrinsic T-cell features significantly influence the outcomes of CAR-T cell therapy. Herein, we review progressing research on T-cell characteristics that impact the effectiveness of CAR-T cells, including T-cell exhaustion, memory subsets, senescence, regulatory T-cells, the CD4+ to CD8+ T-cell ratio, metabolism, and the T-cell receptor repertoire. With comprehensive insight into the biological processes underlying successful CAR-T cell therapy, we will further refine the applications of these novel therapeutic modalities, and enhance their efficacy and safety for patients.
Collapse
Affiliation(s)
- Zhongfei Tao
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Zuzana Chyra
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Jana Kotulová
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Piotr Celichowski
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Jana Mihályová
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Sandra Charvátová
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Roman Hájek
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic.
| |
Collapse
|
4
|
Um IG, Woo JS, Lee YJ, Lee SY, Jeong HY, Na HS, Lee JS, Lee AR, Park SH, Cho ML. IL-21 drives skin and lung inflammation and fibrosis in a model for systemic sclerosis. Immunol Lett 2024; 270:106924. [PMID: 39260526 DOI: 10.1016/j.imlet.2024.106924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Systemic sclerosis (SSc) is an autoimmune disease characterized by vasculopathy, abnormal inflammation, and fibrosis of the skin and internal organs, notably the skin and lungs, significantly impairing quality of life. There is currently no cure for SSc, and its etiology remains largely unknown, presenting a primary barrier to effective treatment. We investigated the role of interleukin-21 (IL-21) in the pathogenesis of SSc. METHODS We assessed the expression levels of fibrosis-related genes in human dermal fibroblasts exposed to IL-21 and TGF beta. We also induced SSc in wild-type C57BL/6 mice and IL-21 knockout (KO) mice with a C57BL/6 background using bleomycin (Bleomycin). Histological analyses were conducted on skin and lung tissues from these mice. The distribution and expression levels of fibrosis-related proteins in the tissues were examined via immunohistochemistry and quantitative real-time PCR. Furthermore, we measured the frequency of Th1, Th2, and Th17 cells among splenocytes through flow cytometry. RESULTS IL-21 activation led to STAT3 phosphorylation more than TGF beta in dermal fibroblasts. In IL-21 KO mice with BLM-induced SSc, skin thickness and lung fibrosis were reduced. The absence of IL-21 in these mice resulted in suppressed expression of fibrosis-related genes, including Col1a1, Col1a2, Col3a1, CTGF, α-SMA, STAT3, and TGFβ, in the skin and lungs. It also led to a decreased frequency of Th1, Th2, and Th17 cells, as well as a lower Th17/Treg ratio among splenocytes, factors known to contribute to the development of SSc. CONCLUSIONS IL-21 contributes to the development of SSc by promoting the expression of fibrosis-related genes and modulating the levels of CD4+ T cells.
Collapse
Affiliation(s)
- In Gyu Um
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin Seok Woo
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Young Joon Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seon-Yeong Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Ha Yeon Jeong
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hun Sik Na
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jeong Su Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - A Ram Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Mi-La Cho
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea.
| |
Collapse
|
5
|
Dong S, Fu C, Shu C, Xie M, Li Y, Zou J, Meng YZ, Xu P, Shan YH, Tian HM, He J, Yang YG, Hu Z. Development of a humanized mouse model with functional human materno-fetal interface immunity. JCI Insight 2024; 9:e176527. [PMID: 39435662 PMCID: PMC11529984 DOI: 10.1172/jci.insight.176527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 09/04/2024] [Indexed: 10/23/2024] Open
Abstract
Materno-fetal immunity possesses specialized characteristics to ensure pathogen clearance while maintaining tolerance to the semiallogeneic fetus. Most of our understanding on human materno-fetal immunity is based on conventional rodent models that may not precisely represent human immunological processes owing to the huge evolutionary divergence. Herein, we developed a pregnant human immune system (HIS) mouse model through busulfan preconditioning, which hosts multilineage human immune subset reconstitution at the materno-fetal interface. Human materno-fetal immunity exhibits a tolerogenic feature at the midgestation stage (embryonic day [E] 14.5), and human immune regulatory subsets were detected in the decidua. However, the immune system switches to an inflammatory profile at the late gestation stage (E19). A cell-cell interaction network contributing to the alternations in the human materno-fetal immune atmosphere was revealed based on single-cell RNA-Seq analysis, wherein human macrophages played crucial roles by secreting several immune regulatory mediators. Furthermore, depletion of Treg cells at E2.5 and E5.5 resulted in severe inflammation and fetus rejection. Collectively, these results demonstrate that the pregnant HIS mouse model permits the development of functional human materno-fetal immunity and offers a tool for human materno-fetal immunity investigation to facilitate drug discovery for reproductive disorders.
Collapse
Affiliation(s)
- Shuai Dong
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Cong Fu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Chang Shu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
| | - Min Xie
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Yan Li
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Jun Zou
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Yi-Zi Meng
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Peng Xu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Yan-Hong Shan
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
| | - Hui-Min Tian
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Jin He
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Zheng Hu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| |
Collapse
|
6
|
Espinoza-García N, Salazar-Camarena DC, Marín-Rosales M, Reyes-Mata MP, Ramírez-Dueñas MG, Muñoz-Valle JF, Borunda-Calderón IM, González-Palacios A, Palafox-Sánchez CA. High Interleukin 21 Levels in Patients with Systemic Lupus Erythematosus: Association with Clinical Variables and rs2221903 Polymorphism. J Clin Med 2024; 13:4512. [PMID: 39124778 PMCID: PMC11313274 DOI: 10.3390/jcm13154512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Background: Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by autoantibody production and diverse tissue and organ inflammatory affections. Interleukin 21 (IL-21) is implicated in B cell survival, proliferation, differentiation, class switching, and immunoglobulin production; therefore, it is considered a key cytokine in the pathogenesis of SLE. However, its association with disease activity and clinical phenotypes remains unclear. We aimed to evaluate the association of IL-21 levels with the disease activity and clinical phenotypes in patients with SLE. Also, we analyzed the IL21 polymorphisms associated with increased IL-21 levels. Methods: The IL-21 serum levels were determined using the enzyme-linked immunosorbent assay (ELISA) method. The rs2221903 and rs2055979 polymorphisms were assessed in 300 healthy controls (HCs) and 300 patients with SLE by the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) technique. The levels of IL-21 were monitored during follow-up visits in 59 patients with SLE. Results: The patients with SLE showed higher IL-21 levels compared to the HCs. The IL-21 levels did not correlate with Mex-SLEDAI and were not different in patients with inactive, mild-moderate, and severe disease. The IL-21 levels were increased in patients with hematological affection. The ROC curve analysis revealed that the IL-21 levels had good predictive power in discriminating among patients with SLE and HCs. In a follow-up analysis, the levels of IL-21 remained higher in the patients with SLE even when the patients were in remission. Also, the rs2221903 polymorphism was associated with increased IL-21 levels. Conclusions: This study highlights the importance of IL-21 as a key cytokine in SLE. IL-21 levels are higher in patients with SLE and remain increased regardless of disease activity. According to the ROC analysis, IL-21 is a potential biomarker of SLE. Further longitudinal studies are needed to explore the relationship between IL-21 and the clinical phenotypes of SLE.
Collapse
Affiliation(s)
- Noemí Espinoza-García
- Doctorado en Ciencias en Biología Molecular en Medicina (DCBMM), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico;
| | - Diana Celeste Salazar-Camarena
- Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (D.C.S.-C.); (M.M.-R.); (M.P.R.-M.); (A.G.-P.)
| | - Miguel Marín-Rosales
- Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (D.C.S.-C.); (M.M.-R.); (M.P.R.-M.); (A.G.-P.)
- Hospital General de Occidente, Secretaría de Salud Jalisco, Guadalajara 45170, Jalisco, Mexico
| | - María Paulina Reyes-Mata
- Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (D.C.S.-C.); (M.M.-R.); (M.P.R.-M.); (A.G.-P.)
| | - María Guadalupe Ramírez-Dueñas
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.G.R.-D.); (J.F.M.-V.)
| | - José Francisco Muñoz-Valle
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.G.R.-D.); (J.F.M.-V.)
| | - Itzel María Borunda-Calderón
- Doctorado en Ciencias Biomédicas (DCB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico;
| | - Aarón González-Palacios
- Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (D.C.S.-C.); (M.M.-R.); (M.P.R.-M.); (A.G.-P.)
| | - Claudia Azucena Palafox-Sánchez
- Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (D.C.S.-C.); (M.M.-R.); (M.P.R.-M.); (A.G.-P.)
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.G.R.-D.); (J.F.M.-V.)
| |
Collapse
|
7
|
Casey M, Lee C, Hoyte SM, Johnston RL, Kwok WY, Law SC, Gandhi MK, Harrison SJ, Nakamura K. Harnessing the cytotoxic granule exocytosis to augment the efficacy of T-cell-engaging bispecific antibody therapy. Haematologica 2024; 109:2131-2143. [PMID: 38268493 PMCID: PMC11215359 DOI: 10.3324/haematol.2023.284435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024] Open
Abstract
T-cell-engaging bispecific antibody (T-BsAb, also known as BiTE) therapy has emerged as a powerful therapeutic modality against multiple myeloma. Given that T-BsAb therapy redirects endogenous T cells to eliminate tumor cells, reinvigorating dysfunctional T cells may be a potential approach to improve the efficacy of T-BsAb. While various immunostimulatory cytokines can potentiate effector T-cell functions, the optimal cytokine treatment for T-BsAb therapy is yet to be established, partly due to a concern of cytokine release syndrome driven by aberrant interferon (IFN)-γ production. Here, we functionally screen immunostimulatory cytokines to determine an ideal combination partner for T-BsAb therapy. This approach reveals interleukin (IL)-21 as a potential immunostimulatory cytokine with the ability to augment T-BsAb-mediated release of granzyme B and perforin, without increasing IFN-γ release. Transcriptome profiling and functional characterization strongly support that IL-21 selectively targets the cytotoxic granule exocytosis pathway, but not pro-inflammatory responses. Notably, IL-21 modulates multiple steps of cytotoxic effector functions including upregulation of co-activating CD226 receptor, increasing cytotoxic granules, and promoting cytotoxic granule delivery at the immunological synapse. Indeed, T-BsAb-mediated myeloma killing is cytotoxic granule-dependent, and IL-21 priming significantly augments cytotoxic activities. Furthermore, in vivo IL-21 treatment induces cytotoxic effector reprogramming in bone marrow T cells, showing synergistic anti-myeloma effects in combination with T-BsAb therapy. Together, harnessing the cytotoxic granule exocytosis pathway by IL-21 may be a potential approach to achieve better responses by T-BsAb therapy.
Collapse
Affiliation(s)
- Mika Casey
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD
| | - Carol Lee
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD
| | - Sharon M Hoyte
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD
| | - Rebecca L Johnston
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD
| | - Wing Yu Kwok
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD
| | - Soi Cheng Law
- Mater Research, University of Queensland, Brisbane, QLD
| | | | - Simon J Harrison
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital Melbourne VIC Australia; Sir Peter MacCallum, Department of Oncology, University of Melbourne, Parkville
| | - Kyohei Nakamura
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD.
| |
Collapse
|
8
|
Jones PW, Mallat Z, Nus M. T-Cell/B-Cell Interactions in Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:1502-1511. [PMID: 38813700 PMCID: PMC11208060 DOI: 10.1161/atvbaha.124.319845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Atherosclerosis is a complex inflammatory disease in which the adaptive immune response plays an important role. While the overall impact of T and B cells in atherosclerosis is relatively well established, we are only beginning to understand how bidirectional T-cell/B-cell interactions can exert prominent atheroprotective and proatherogenic functions. In this review, we will focus on these T-cell/B-cell interactions and how we could use them to therapeutically target the adaptive immune response in atherosclerosis.
Collapse
Affiliation(s)
- Peter William Jones
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute, University of Cambridge, United Kingdom (P.W.J., Z.M., M.N.)
| | - Ziad Mallat
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute, University of Cambridge, United Kingdom (P.W.J., Z.M., M.N.)
- INSERM U970, Paris Cardiovascular Research Centre, France (Z.M.)
| | - Meritxell Nus
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute, University of Cambridge, United Kingdom (P.W.J., Z.M., M.N.)
| |
Collapse
|
9
|
Li Y, Ramírez-Suástegui C, Harris R, Castañeda-Castro FE, Ascui G, Pérez-Jeldres T, Diaz A, Morong C, Giles DA, Chai J, Seumois G, Sanchez-Elsner T, Cummings F, Kronenberg M, Vijayanand P. Stem-like T cells are associated with the pathogenesis of ulcerative colitis in humans. Nat Immunol 2024; 25:1231-1244. [PMID: 38898157 DOI: 10.1038/s41590-024-01860-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 05/01/2024] [Indexed: 06/21/2024]
Abstract
To understand the role of T cells in the pathogenesis of ulcerative colitis (UC), we analyzed colonic T cells isolated from patients with UC and controls. Here we identified colonic CD4+ and CD8+ T lymphocyte subsets with gene expression profiles resembling stem-like progenitors, previously reported in several mouse models of autoimmune disease. Stem-like T cells were increased in inflamed areas compared to non-inflamed regions from the same patients. Furthermore, TCR sequence analysis indicated stem-like T cells were clonally related to proinflammatory T cells, suggesting their involvement in sustaining effectors that drive inflammation. Using an adoptive transfer colitis model in mice, we demonstrated that CD4+ T cells deficient in either BCL-6 or TCF1, transcription factors that promote T cell stemness, had decreased colon T cells and diminished pathogenicity. Our results establish a strong association between stem-like T cell populations and UC pathogenesis, highlighting the potential of targeting this population to improve clinical outcomes.
Collapse
Affiliation(s)
- Yingcong Li
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Richard Harris
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Gabriel Ascui
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Tamara Pérez-Jeldres
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Gastroenterology, Hospital San Borja Arriarán, Santiago, Chile
| | - Alejandro Diaz
- Department of Gastroenterology, Hospital San Borja Arriarán, Santiago, Chile
| | - Carla Morong
- Department of Gastroenterology, Hospital San Borja Arriarán, Santiago, Chile
| | - Daniel A Giles
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Lineage Therapeutics, Carlsbad, CA, USA
| | - Jiani Chai
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Pathology, Albert Einstein Medical College, New York, NY, USA
| | | | - Tilman Sanchez-Elsner
- Department of Gastroenterology, University Hospital Southampton NHS FT, Southampton, UK
| | - Fraser Cummings
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Department of Gastroenterology, University Hospital Southampton NHS FT, Southampton, UK
| | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Molecular Biology, University of California, San Diego, San Diego, CA, USA.
| | - Pandurangan Vijayanand
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Medicine, University of California, San Diego, San Diego, CA, USA.
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
10
|
Shufang M, Xiaojiao H, Yinhong K. Pro-inflammatory cytokine IL-21 correlates with the reactive oxygen species and 25-hydroxy vitamin D in rheumatoid arthritis patients. Immun Inflamm Dis 2024; 12:e1308. [PMID: 39056553 PMCID: PMC11273535 DOI: 10.1002/iid3.1308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/28/2024] [Accepted: 05/19/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disorder and its characteristics include the immune system's invasion of the healthy lining of the joints and the articular structures degeneration. The IL-21 pro-inflammatory cytokine, and the reactive oxygen species (ROS) might have a role in the RA etiopathogenesis. The present study assessed the correlation of IL-21 with vitamin 25(OH)D and the ROS. METHODS The study included 120 RA patients and 60 healthy group. The RA patients were categorized based on rheumatoid factor (RF) seropositivity or seronegativity and the RA severity. Chemiluminescent immunoassay and 10% hematocrit were used to check levels of vitamin 25(OH)D and ROS, respectively. ELISA was used for the detection of IL-21 in the plasma. RESULTS The RA patients had a significantly reduced vitamin 25(OH)D level compared to the healthy controls. The IL-21 and ROS were however significantly increased in the RA patients compared to the controls. Further, the seropositive RF and the high RA severity patients had significant IL-21 and ROS increase in comparison with the seronegative RF and the low severity RA patients. Finally, IL-21 negatively correlated with vitamin 25(OH)D, but positively correlated with the ROS. CONCLUSION This is the first investigation to confirm the relationship between IL-21 with vitamin 25(OH)D and the ROS among the RA patients. The findings indicate that vitamin 25(OH)D is reduced in the RA patients' serum. ROS and IL-21 are also associated with increased RA severity.
Collapse
Affiliation(s)
- Ma Shufang
- Rheumatology and Immunology DepartmentFourth Central Hospital of Baoding CityBaodingHebei ProvinceChina
| | - Han Xiaojiao
- Rheumatology and Immunology DepartmentFourth Central Hospital of Baoding CityBaodingHebei ProvinceChina
| | - Kang Yinhong
- Obstetrics DepartmentFourth Central Hospital of Baoding CityBaodingHebei ProvinceChina
| |
Collapse
|
11
|
Tatomir A, Anselmo F, Boodhoo D, Chen H, Mekala AP, Nguyen V, Cuevas J, Rus V, Rus H. Multiple sclerosis disease activity, a multi-biomarker score of disease activity and response to treatment in multiple sclerosis. Front Immunol 2024; 15:1338585. [PMID: 38994359 PMCID: PMC11236682 DOI: 10.3389/fimmu.2024.1338585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
Regular assessment of disease activity in relapsing-remitting multiple sclerosis (RRMS) is required to optimize clinical outcomes. Biomarkers can be a valuable tool for measuring disease activity in multiple sclerosis (MS) if they reflect the pathological processes underlying MS pathogenicity. In this pilot study, we combined multiple biomarkers previously analyzed in RRMS patients into an MS disease activity (MSDA) score to evaluate their ability to predict relapses and treatment response to glatiramer acetate (GA). Response Gene to Complement 32 (RGC-32), FasL, IL-21, SIRT1, phosphorylated SIRT1 (p-SIRT1), and JNK1 p54 levels were used to generate cut-off values for each biomarker. Any value below the cutoff for RGC-32, FasL SIRT1, or p-SIRT1 or above the cutoff for IL-21 or JNK1 p54 was given a +1 value, indicating relapse or lack of response to GA. Any value above the cutoff value for RGC-32, FasL, SIRT1, p-SIRT1 or below that for IL-21 or JNK1 p54 was given a -1 value, indicating clinical stability or response to GA. An MSDA score above +1 indicated a relapse or lack of response to treatment. An MSDA score below -1 indicated clinical stability or response to treatment. Our results showed that the MSDA scores generated using either four or six biomarkers had a higher sensitivity and specificity and significantly correlated with the expanded disability status scale. Although these results suggest that the MSDA test can be useful for monitoring therapeutic response to biologic agents and assessing clinically challenging situations, the present findings need to be confirmed in larger studies.
Collapse
Affiliation(s)
- Alexandru Tatomir
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Neurology Department, Baltimore Veterans Administration Hospital, Baltimore, MD, United States
| | - Freidrich Anselmo
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Dallas Boodhoo
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Hegang Chen
- Department of Epidemiology and Public Health, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Armugam P. Mekala
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Vinh Nguyen
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Jacob Cuevas
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Horea Rus
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Neurology Department, Baltimore Veterans Administration Hospital, Baltimore, MD, United States
| |
Collapse
|
12
|
Malahe SRK, den Hartog Y, Rietdijk WJR, van Baarle D, de Kuiper R, Reijerkerk D, Ras AM, Geers D, Diavatopoulos DA, Messchendorp AL, van der Molen RG, Imhof C, Frölke SC, Bemelman FJ, Gansevoort RT, Hilbrands LB, Sanders JSF, GeurtsvanKessel CH, Kho MML, de Vries RD, Reinders MEJ, Baan CC. Repeated COVID-19 Vaccination Drives Memory T- and B-cell Responses in Kidney Transplant Recipients: Results From a Multicenter Randomized Controlled Trial. Transplantation 2024; 108:00007890-990000000-00797. [PMID: 38902860 PMCID: PMC11581438 DOI: 10.1097/tp.0000000000005119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/30/2024] [Accepted: 05/16/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Insight into cellular immune responses to COVID-19 vaccinations is crucial for optimizing booster programs in kidney transplant recipients (KTRs). METHODS In an immunologic substudy of a multicenter randomized controlled trial (NCT05030974) investigating different repeated vaccination strategies in KTR who showed poor serological responses after 2 or 3 doses of an messenger RNA (mRNA)-based vaccine, we compared SARS-CoV-2-specific interleukin-21 memory T-cell and B-cell responses by enzyme-linked immunosorbent spot (ELISpot) assays and serum IgG antibody levels. Patients were randomized to receive: a single dose of mRNA-1273 (100 μg, n = 25), a double dose of mRNA-1273 (2 × 100 μg, n = 25), or a single dose of adenovirus type 26 encoding the SARS-CoV-2 spike glycoprotein (Ad26.COV2.S) (n = 25). In parallel, we also examined responses in 50 KTR receiving 100 μg mRNA-1273, randomized to continue (n = 25) or discontinue (n = 25) mycophenolate mofetil/mycophenolic acid. As a reference, the data were compared with KTR who received 2 primary mRNA-1273 vaccinations. RESULTS Repeated vaccination increased the seroconversion rate from 21% to 66% in all patients, which was strongly associated with enhanced levels of SARS-CoV-2-specific interleukin-21 memory T cells (odd ratio, 3.84 [1.89-7.78]; P < 0.001) and B cells (odd ratio, 35.93 [6.94-186.04]; P < 0.001). There were no significant differences observed in these responses among various vaccination strategies. In contrast to KTR vaccinated with 2 primary vaccinations, the number of antigen-specific memory B cells demonstrated potential for classifying seroconversion after repeated vaccination (area under the curve, 0.64; 95% confidence interval, 0.37-0.90; P = 0.26 and area under the curve, 0.95; confidence interval, 0.87-0.97; P < 0.0001, respectively). CONCLUSIONS Our study emphasizes the importance of virus-specific memory T- and B-cell responses for comprehensive understanding of COVID-19 vaccine efficacy among KTR.
Collapse
Affiliation(s)
- S. Reshwan K. Malahe
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Yvette den Hartog
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Wim J. R. Rietdijk
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, Virology and Immunology Research Group, University Medical Center Groningen, Groningen, the Netherlands
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Ronella de Kuiper
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Derek Reijerkerk
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alicia M. Ras
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Daryl Geers
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Dimitri A. Diavatopoulos
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, the Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center Nijmegen, Nijmegen, the Netherlands
| | - A. Lianne Messchendorp
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Renate G. van der Molen
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, the Netherlands
| | - Céline Imhof
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sophie C. Frölke
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Frederike J. Bemelman
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Ron T. Gansevoort
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Luuk B. Hilbrands
- Department of Nephrology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jan-Stephan F. Sanders
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Marcia M. L. Kho
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marlies E. J. Reinders
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Carla C. Baan
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
13
|
Johnson JT, Surette FA, Ausdahl GR, Shah M, Minns AM, Lindner SE, Zander RA, Butler NS. CD4 T Cell-Derived IL-21 Is Critical for Sustaining Plasmodium Infection-Induced Germinal Center Responses and Promoting the Selection of Memory B Cells with Recall Potential. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1467-1478. [PMID: 38477614 PMCID: PMC11018477 DOI: 10.4049/jimmunol.2300683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/25/2024] [Indexed: 03/14/2024]
Abstract
Development of Plasmodium-specific humoral immunity is critically dependent on CD4 Th cell responses and germinal center (GC) reactions during blood-stage Plasmodium infection. IL-21, a cytokine primarily produced by CD4 T cells, is an essential regulator of affinity maturation, isotype class-switching, B cell differentiation, and maintenance of GC reactions in response to many infection and immunization models. In models of experimental malaria, mice deficient in IL-21 or its receptor IL-21R fail to develop memory B cell populations and are not protected against secondary infection. However, whether sustained IL-21 signaling in ongoing GCs is required for maintaining GC magnitude, organization, and output is unclear. In this study, we report that CD4+ Th cells maintain IL-21 expression after resolution of primary Plasmodium yoelii infection. We generated an inducible knockout mouse model that enabled cell type-specific and timed deletion of IL-21 in peripheral, mature CD4 T cells. We found that persistence of IL-21 signaling in active GCs had no impact on the magnitude of GC reactions or their capacity to produce memory B cell populations. However, the memory B cells generated in the absence of IL-21 exhibited reduced recall function upon challenge. Our data support that IL-21 prevents premature cellular dissolution within the GC and promotes stringency of selective pressures during B cell fate determination required to produce high-quality Plasmodium-specific memory B cells. These data are additionally consistent with a temporal requirement for IL-21 in fine-tuning humoral immune memory responses during experimental malaria.
Collapse
Affiliation(s)
- Jordan T. Johnson
- Graduate Program in Immunology, University of Iowa, Iowa City, Iowa USA
- These authors contributed equally
| | - Fionna A. Surette
- Graduate Program in Immunology, University of Iowa, Iowa City, Iowa USA
- These authors contributed equally
| | - Graham R. Ausdahl
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa USA
| | - Manan Shah
- Graduate Program in Immunology, University of Iowa, Iowa City, Iowa USA
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa USA
| | - Allen M. Minns
- Department of Biochemistry & Molecular Biology, Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania USA
| | - Scott E. Lindner
- Department of Biochemistry & Molecular Biology, Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania USA
| | - Ryan A. Zander
- Graduate Program in Immunology, University of Iowa, Iowa City, Iowa USA
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa USA
| | - Noah S. Butler
- Graduate Program in Immunology, University of Iowa, Iowa City, Iowa USA
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa USA
| |
Collapse
|
14
|
Zhu W, Zhang Z, Chen J, Chen X, Huang L, Zhang X, Huang X, Ma N, Xu W, Yi X, Lu X, Fu X, Li S, Mo G, Wang Y, Yuan G, Zang M, Li Q, Jiang X, He Y, Wu S, He Y, Li Y, Hou J. A novel engineered IL-21 receptor arms T-cell receptor-engineered T cells (TCR-T cells) against hepatocellular carcinoma. Signal Transduct Target Ther 2024; 9:101. [PMID: 38643203 PMCID: PMC11032311 DOI: 10.1038/s41392-024-01792-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 01/30/2024] [Accepted: 03/07/2024] [Indexed: 04/22/2024] Open
Abstract
Strategies to improve T cell therapy efficacy in solid tumors such as hepatocellular carcinoma (HCC) are urgently needed. The common cytokine receptor γ chain (γc) family cytokines such as IL-2, IL-7, IL-15 and IL-21 play fundamental roles in T cell development, differentiation and effector phases. This study aims to determine the combination effects of IL-21 in T cell therapy against HCC and investigate optimized strategies to utilize the effect of IL-21 signal in T cell therapy. The antitumor function of AFP-specific T cell receptor-engineered T cells (TCR-T) was augmented by exogenous IL-21 in vitro and in vivo. IL-21 enhanced proliferation capacity, promoted memory differentiation, downregulated PD-1 expression and alleviated apoptosis in TCR-T after activation. A novel engineered IL-21 receptor was established, and TCR-T armed with the novel engineered IL-21 receptors (IL-21R-TCR-T) showed upregulated phosphorylated STAT3 expression without exogenous IL-21 ligand. IL-21R-TCR-T showed better proliferation upon activation and superior antitumor function in vitro and in vivo. IL-21R-TCR-T exhibited a less differentiated, exhausted and apoptotic phenotype than conventional TCR-T upon repetitive tumor antigen stimulation. The novel IL-21 receptor in our study programs powerful TCR-T and can avoid side effects induced by IL-21 systemic utilization. The novel IL-21 receptor creates new opportunities for next-generation TCR-T against HCC.
Collapse
Affiliation(s)
- Wei Zhu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiming Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinzhang Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaolan Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Huang
- Institute of Cellular Medicine, Newcastle University Medical School, Newcastle, UK
| | - Xiaoyong Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuan Huang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Na Ma
- Department of Pathology, The First People's Hospital of Foshan, Foshan, China
| | - Weikang Xu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuan Yi
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Xinyu Lu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Fu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Siwei Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guoheng Mo
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyue Wang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guosheng Yuan
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengya Zang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qi Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaotao Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yajing He
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sha Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Key Laboratory of Proteomics of Guangdong Province, Demonstration Center for Experimental Education of Basic Medical Sciences of China, Guangzhou, China
| | - Yukai He
- Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, USA
| | - Yongyin Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Jinlin Hou
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
15
|
Isvoranu G, Chiritoiu-Butnaru M. Therapeutic potential of interleukin-21 in cancer. Front Immunol 2024; 15:1369743. [PMID: 38638431 PMCID: PMC11024325 DOI: 10.3389/fimmu.2024.1369743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/12/2024] [Indexed: 04/20/2024] Open
Abstract
Interleukin-21 (IL-21) is an immunostimulatory cytokine which belongs to the common gamma-chain family of cytokines. It plays an import role in the development, differentiation, proliferation, and activation of immune cells, in particular T and natural killer (NK) cells. Since its discovery in 2000, IL-21 has been shown to regulate both adaptive and immune responses associates with key role in antiviral and antitumor responses. Recent advances indicate IL-21 as a promising target for cancer treatment and encouraging results were obtained in preclinical studies which investigated the potency of IL-21 alone or in combination with other therapies, including monoclonal antibodies, checkpoint inhibitory molecules, oncolytic virotherapy, and adoptive cell transfer. Furthermore, IL-21 showed antitumor effects in the treatment of patients with advanced cancer, with minimal side effects in several clinical trials. In the present review, we will outline the recent progress in IL-21 research, highlighting the potential of IL-21 based therapy as single agent or in combination with other drugs to enhance cancer treatment efficiency.
Collapse
Affiliation(s)
- Gheorghita Isvoranu
- Department of Animal Husbandry,” Victor Babeș” National Institute of Pathology, Bucharest, Romania
| | - Marioara Chiritoiu-Butnaru
- Department of Molecular and Cell Biology, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| |
Collapse
|
16
|
Zhang X, Hu Q, Jiang B, Xie F, Zhang Z, Hafezi-Moghadam A, Sun D. Role of Interleukin-21 in retinal ischemia-reperfusion injury: Unveiling the impact on retinal ganglion cell apoptosis. Int Immunopharmacol 2024; 128:111480. [PMID: 38194747 DOI: 10.1016/j.intimp.2023.111480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/22/2023] [Accepted: 12/30/2023] [Indexed: 01/11/2024]
Abstract
BACKGROUND Retinal ischemia-reperfusion (I/R) serves as a significant contributor to ocular diseases, triggering a cascade of pathological processes. The interplay between neuroinflammation and the apoptosis of retinal ganglion cell (RGC) is a well-explored aspect of retinal I/R-induced tissue damage. Within this intricate landscape, the inflammatory cytokine Interleukin-21 (IL21) emerges as a potent mediator of neuroinflammation with known detrimental effects on neuronal integrity. However, its specific impact on RGC apoptosis in the context of retinal I/R has remains to be uncovered. This study aims to unravel the potential anti-apoptotic effects of IL21 siRNA on RGC, shedding light on the neuroprotection of retinal I/R. METHODS Sprague-Dawley (SD) rats underwent a controlled elevation of intraocular pressure (IOP) to 110 mmHg for 60 min to simulate retinal I/R conditions. To explore the influence of IL21 on RGC apoptosis and its underlying molecular mechanisms, a comprehensive array of techniques such immunohistochemistry, immunofluorescence, TUNEL, Hematoxylin-eosin (H&E), immunoblotting, and qRT-PCR were carried out. RESULTS The landscape of retinal I/R injury revealed an increase in the expression of IL21, reaching its peak at 72 h. Notably, IL21 markedly induced RGC apoptosis within the retinal I/R milieu. The introduction of IL21 siRNA showed promising outcomes, manifesting as an amelioration of neurological function deficits, a reduction in RGC loss, and an increase in the thickness of the inner retinal layer at the 72-hour reperfusion. Additionally, IL21 siRNA demonstrated its ability to hinder the release of proteins associated with apoptosis via the JAK/STAT signaling pathway. In the in vitro setting, IL21 siRNA efficiently reduced R28 cell apoptosis by suppressing the production of proteins associated with apoptosis by regulating the JAK/STAT signaling pathway. CONCLUSIONS This study provides evidence for the pathogenic role of IL21 in retinal I/R. The findings underscore IL21 siRNA as a promising therapeutic target for ischemic retinal injury. Its efficacy lies in its ability to mitigate RGC apoptosis by suppressing the JAK/STAT signaling pathway. These findings not only enhance our comprehension of retinal I/R pathology but also suggests IL21 siRNA as a potential transformative factor in the development of targeted therapies for ischemic retinal injuries.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150086, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Qiang Hu
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150086, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Bo Jiang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Fang Xie
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Zhongyu Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ali Hafezi-Moghadam
- Molecular Biomarkers Nano-Imaging Laboratory (MBNI), Brigham & Women's Hospital, Boston, MA 02115, USA
| | - Dawei Sun
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150086, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China.
| |
Collapse
|
17
|
Schramm CA, Moon D, Peyton L, Lima NS, Wake C, Boswell KL, Henry AR, Laboune F, Ambrozak D, Darko SW, Teng IT, Foulds KE, Carfi A, Edwards DK, Kwong PD, Koup RA, Seder RA, Douek DC. Interaction dynamics between innate and adaptive immune cells responding to SARS-CoV-2 vaccination in non-human primates. Nat Commun 2023; 14:7961. [PMID: 38042809 PMCID: PMC10693617 DOI: 10.1038/s41467-023-43420-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/08/2023] [Indexed: 12/04/2023] Open
Abstract
As SARS-CoV-2 variants continue evolving, testing updated vaccines in non-human primates remains important for guiding human clinical practice. To date, such studies have focused on antibody titers and antigen-specific B and T cell frequencies. Here, we extend our understanding by integrating innate and adaptive immune responses to mRNA-1273 vaccination in rhesus macaques. We sorted innate immune cells from a pre-vaccine time point, as well as innate immune cells and antigen-specific peripheral B and T cells two weeks after each of two vaccine doses and used single-cell sequencing to assess the transcriptomes and adaptive immune receptors of each cell. We show that a subset of S-specific T cells expresses cytokines critical for activating innate responses, with a concomitant increase in CCR5-expressing intermediate monocytes and a shift of natural killer cells to a more cytotoxic phenotype. The second vaccine dose, administered 4 weeks after the first, elicits an increase in circulating germinal center-like B cells 2 weeks later, which are more clonally expanded and enriched for epitopes in the receptor binding domain. Both doses stimulate inflammatory response genes associated with elevated antibody production. Overall, we provide a comprehensive picture of bidirectional signaling between innate and adaptive components of the immune system and suggest potential mechanisms for the enhanced response to secondary exposure.
Collapse
Affiliation(s)
- Chaim A Schramm
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Damee Moon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lowrey Peyton
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Noemia S Lima
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Christian Wake
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kristin L Boswell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Amy R Henry
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Farida Laboune
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David Ambrozak
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Samuel W Darko
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - I-Ting Teng
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kathryn E Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
18
|
Yadav M, Uikey BN, Rathore SS, Gupta P, Kashyap D, Kumar C, Shukla D, Vijayamahantesh, Chandel AS, Ahirwar B, Singh AK, Suman SS, Priyadarshi A, Amit A. Role of cytokine in malignant T-cell metabolism and subsequent alternation in T-cell tumor microenvironment. Front Oncol 2023; 13:1235711. [PMID: 37746258 PMCID: PMC10513393 DOI: 10.3389/fonc.2023.1235711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
T cells are an important component of adaptive immunity and T-cell-derived lymphomas are very complex due to many functional sub-types and functional elasticity of T-cells. As with other tumors, tissues specific factors are crucial in the development of T-cell lymphomas. In addition to neoplastic cells, T- cell lymphomas consist of a tumor micro-environment composed of normal cells and stroma. Numerous studies established the qualitative and quantitative differences between the tumor microenvironment and normal cell surroundings. Interaction between the various component of the tumor microenvironment is crucial since tumor cells can change the microenvironment and vice versa. In normal T-cell development, T-cells must respond to various stimulants deferentially and during these courses of adaptation. T-cells undergo various metabolic alterations. From the stage of quiescence to attention of fully active form T-cells undergoes various stage in terms of metabolic activity. Predominantly quiescent T-cells have ATP-generating metabolism while during the proliferative stage, their metabolism tilted towards the growth-promoting pathways. In addition to this, a functionally different subset of T-cells requires to activate the different metabolic pathways, and consequently, this regulation of the metabolic pathway control activation and function of T-cells. So, it is obvious that dynamic, and well-regulated metabolic pathways are important for the normal functioning of T-cells and their interaction with the microenvironment. There are various cell signaling mechanisms of metabolism are involved in this regulation and more and more studies have suggested the involvement of additional signaling in the development of the overall metabolic phenotype of T cells. These important signaling mediators include cytokines and hormones. The impact and role of these mediators especially the cytokines on the interplay between T-cell metabolism and the interaction of T-cells with their micro-environments in the context of T-cells lymphomas are discussed in this review article.
Collapse
Affiliation(s)
- Megha Yadav
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Blessi N. Uikey
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | | | - Priyanka Gupta
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Diksha Kashyap
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Chanchal Kumar
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Dhananjay Shukla
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Vijayamahantesh
- Department of Immunology and Microbiology, University of Missouri, Columbia, SC, United States
| | - Arvind Singh Chandel
- Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo, Japan
| | - Bharti Ahirwar
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | | | - Shashi Shekhar Suman
- Department of Zoology, Udayana Charya (UR) College, Lalit Narayan Mithila University, Darbhanga, India
| | - Amit Priyadarshi
- Department of Zoology, Veer Kunwar Singh University, Arrah, India
| | - Ajay Amit
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| |
Collapse
|
19
|
Malahe SRK, Hartog YD, Rietdijk WJR, van Baarle D, de Kuiper R, Reijerkerk D, Ras AM, Geers D, Diavatopoulos DA, Messchendorp AL, van der Molen RG, Remmerswaal EBM, Bemelman FJ, Gansevoort RT, Hilbrands LB, Sanders JS, GeurtsvanKessel CH, Kho MML, de Vries RD, Reinders MEJ, Baan CC. The role of interleukin-21 in COVID-19 vaccine-induced B cell-mediated immune responses in patients with kidney disease and kidney transplant recipients. Am J Transplant 2023; 23:1411-1424. [PMID: 37270109 PMCID: PMC10234364 DOI: 10.1016/j.ajt.2023.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 05/02/2023] [Accepted: 05/22/2023] [Indexed: 06/05/2023]
Abstract
T-cell-mediated help to B cells is required for the development of humoral responses, in which the cytokine interleukin (IL)-21 is key. Here, we studied the mRNA-1273 vaccine-induced SARS-CoV-2-specific memory T-cell IL-21 response, memory B cell response, and immunoglobulin (Ig)G antibody levels in peripheral blood at 28 days after the second vaccination by ELISpot and the fluorescent bead-based multiplex immunoassay, respectively. We included 40 patients with chronic kidney disease (CKD), 34 patients on dialysis, 63 kidney transplant recipients (KTR), and 47 controls. We found that KTR, but not patients with CKD and those receiving dialysis, showed a significantly lower number of SARS-CoV-2-specific IL-21 producing T cells than controls (P < .001). KTR and patients with CKD showed lower numbers of SARS-CoV-2-specific IgG-producing memory B cells when compared with controls (P < .001 and P = .01, respectively). The T-cell IL-21 response was positively associated with the SARS-CoV-2-specific B cell response and the SARS-CoV-2 spike S1-specific IgG antibody levels (both Pearson r = 0.5; P < .001). In addition, SARS-CoV-2-specific B cell responses were shown to be IL-21 dependent. Taken together, we show that IL-21 signaling is important in eliciting robust B cell-mediated immune responses in patients with kidney disease and KTR.
Collapse
Affiliation(s)
- S Reshwan K Malahe
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Yvette den Hartog
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Wim J R Rietdijk
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Debbie van Baarle
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Ronella de Kuiper
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Derek Reijerkerk
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Alicia M Ras
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Daryl Geers
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Dimitri A Diavatopoulos
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | - A Lianne Messchendorp
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Renate G van der Molen
- Radboud Institute for Molecular Life Sciences, Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | - Ester B M Remmerswaal
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Frederike J Bemelman
- Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ron T Gansevoort
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jan-Stephan Sanders
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | | | - Marcia M L Kho
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Rory D de Vries
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Marlies E J Reinders
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
20
|
Guo X, Li H, Meng X, Zhao Z, Zhang R, Wang L, Li J. CD8 + T-cell number and function are altered by Shkbp1 knockout mediated suppression of tumor growth in mice. Mol Immunol 2023; 160:32-43. [PMID: 37343421 DOI: 10.1016/j.molimm.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/12/2023] [Accepted: 06/06/2023] [Indexed: 06/23/2023]
Abstract
CD8 + effector cells are highly skilled in immune surveillance and contribute to adaptive immunity against cancer cells. An increasing number of molecular factors affecting T-cell differentiation may alter T-cell function by increasing or decreasing the capacity of the immune system to kill cancer cells. Here, Sh3kbp1 binding protein 1 (Shkbp1), known as CIN85 binding protein or SETA binding protein, was found to be expressed in immune organs and immune cells. Shkbp1 knockout mice presented abnormal red and white pulp structures in spleen. Shkbp1 knockout increased CD8 + T cell number in spleen and enhanced the function of isolated CD8 + T cells from Shkbp1 knockout mice. The subcutaneous melanoma model in Shkbp1 knockout mice showed that tumor growth was inhibited, and the infiltration of CD8 + T cells in tumor tissue was increased. Furthermore, adenoviral therapy targeting Shkbp1 indicated that knockout of Shkbp1 increased CD8 + T cells and inhibited tumor growth. This study provides new insights into the role of Shkbp1 in CD8 differentiation and functions, suggesting that Shkbp1 may be a new, potential target in cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaolan Guo
- Institute of Basic Medical Sciences, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Haobin Li
- Institute of Basic Medical Sciences, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiuqiong Meng
- Institute of Basic Medical Sciences, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - ZhiBin Zhao
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510006, China
| | - Rongxin Zhang
- Institute of Basic Medical Sciences, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lijing Wang
- Institute of Basic Medical Sciences, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Jiangchao Li
- Institute of Basic Medical Sciences, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
21
|
Ildefonso GV, Finley SD. A data-driven Boolean model explains memory subsets and evolution in CD8+ T cell exhaustion. NPJ Syst Biol Appl 2023; 9:36. [PMID: 37524735 PMCID: PMC10390540 DOI: 10.1038/s41540-023-00297-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/04/2023] [Indexed: 08/02/2023] Open
Abstract
T cells play a key role in a variety of immune responses, including infection and cancer. Upon stimulation, naïve CD8+ T cells proliferate and differentiate into a variety of memory and effector cell types; however, failure to clear antigens causes prolonged stimulation of CD8+ T cells, ultimately leading to T cell exhaustion (TCE). The functional and phenotypic changes that occur during CD8+ T cell differentiation are well characterized, but the underlying gene expression state changes are not completely understood. Here, we utilize a previously published data-driven Boolean model of gene regulatory interactions shown to mediate TCE. Our network analysis and modeling reveal the final gene expression states that correspond to TCE, along with the sequence of gene expression patterns that give rise to those final states. With a model that predicts the changes in gene expression that lead to TCE, we could evaluate strategies to inhibit the exhausted state. Overall, we demonstrate that a common pathway model of CD8+ T cell gene regulatory interactions can provide insights into the transcriptional changes underlying the evolution of cell states in TCE.
Collapse
Affiliation(s)
- Geena V Ildefonso
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Stacey D Finley
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA.
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, California, USA.
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
22
|
Aghbash PS, Rasizadeh R, Arefi V, Nahand JS, Baghi HB. Immune-checkpoint expression in antigen-presenting cells (APCs) of cytomegaloviruses infection after transplantation: as a diagnostic biomarker. Arch Microbiol 2023; 205:280. [PMID: 37430000 DOI: 10.1007/s00203-023-03623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/12/2023]
Abstract
Cytomegalovirus (CMV), a member of the Herpesviridae family, mostly causes only slight feverish symptoms or can be asymptomatic in immunocompetent individuals. However, it is known to be particularly a significant cause of morbidity in immunocompromised patients, including transplant recipients, whose immune system has been weakened due to the consumption of immunosuppressor drugs. Therefore, the diagnosis of CMV infection after transplantation is crucial. New diagnostic methods for the quick detection of CMV have been developed as a result of understanding the clinical importance of invasive CMV. Antigen-presenting cells (APCs) and T cells are important components of the immune system and it may be possible to diagnose viral infections using immunological markers, such as lymphocytosis, cytotoxic T lymphocytes (CTL), and serum cytokine levels. Moreover, PD-1, CTLA 4, and TIGIT, which are expressed on certain T cells and antigen-presenting cells, are over-expressed during the infection. The assessment of CMV infection based on T cell and APC activity, and the expression of immunological checkpoints, can be helpful for the diagnosis of transplant patients at risk for CMV infection. In this review, we will investigate how immune checkpoints affect immune cells and how they impair organ transplantation after CMV infection.
Collapse
Affiliation(s)
- Parisa Shiri Aghbash
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reyhaneh Rasizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Arefi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
| | - Hossein Bannazadeh Baghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran.
| |
Collapse
|
23
|
Ciecko AE, Wang Y, Harleston S, Drewek A, Serreze DV, Geurts AM, Lin CW, Chen YG. Heterogeneity of Islet-Infiltrating IL-21+ CD4 T Cells in a Mouse Model of Type 1 Diabetes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:935-946. [PMID: 36762954 PMCID: PMC10483376 DOI: 10.4049/jimmunol.2200712] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/29/2023] [Indexed: 02/11/2023]
Abstract
IL-21 is essential for type 1 diabetes (T1D) development in the NOD mouse model. IL-21-expressing CD4 T cells are present in pancreatic islets where they contribute to T1D progression. However, little is known about their phenotype and differentiation states. To fill this gap, we generated, to our knowledge, a novel IL-21 reporter NOD strain to further characterize IL-21+ CD4 T cells in T1D. IL-21+ CD4 T cells accumulate in pancreatic islets and recognize β cell Ags. Single-cell RNA sequencing revealed that CD4 T effector cells in islets actively express IL-21 and they are highly diabetogenic despite expressing multiple inhibitory molecules, including PD-1 and LAG3. Islet IL-21+ CD4 T cells segregate into four phenotypically and transcriptionally distinct differentiation states, that is, less differentiated early effectors, T follicular helper (Tfh)-like cells, and two Th1 subsets. Trajectory analysis predicts that early effectors differentiate into both Tfh-like and terminal Th1 cells. We further demonstrated that intrinsic IL-27 signaling controls the differentiation of islet IL-21+ CD4 T cells, contributing to their helper function. Collectively, our study reveals the heterogeneity of islet-infiltrating IL-21+ CD4 T cells and indicates that both Tfh-like and Th1 subsets produce IL-21 throughout their differentiation process, highlighting the important sources of IL-21 in T1D pathogenesis.
Collapse
Affiliation(s)
- Ashley E Ciecko
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
- The Max McGee Research Center for Juvenile Diabetes, Children's Research Institute of Children's Hospital of Wisconsin, Milwaukee, WI
| | - Yu Wang
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI
| | - Stephanie Harleston
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
- The Max McGee Research Center for Juvenile Diabetes, Children's Research Institute of Children's Hospital of Wisconsin, Milwaukee, WI
| | - Amber Drewek
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
- The Max McGee Research Center for Juvenile Diabetes, Children's Research Institute of Children's Hospital of Wisconsin, Milwaukee, WI
| | | | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI
| | - Chien-Wei Lin
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI
| | - Yi-Guang Chen
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
- The Max McGee Research Center for Juvenile Diabetes, Children's Research Institute of Children's Hospital of Wisconsin, Milwaukee, WI
| |
Collapse
|
24
|
Brynedal B, Yoosuf N, Ulfarsdottir TB, Ziemek D, Maciejewski M, Folkersen L, Westerlind H, Müller M, Sahlström P, Jelinsky SA, Hensvold A, Padyukov L, Pomiano NV, Catrina A, Klareskog L, Berg L. Molecular signature of methotrexate response among rheumatoid arthritis patients. Front Med (Lausanne) 2023; 10:1146353. [PMID: 37051216 PMCID: PMC10084884 DOI: 10.3389/fmed.2023.1146353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/22/2023] [Indexed: 03/29/2023] Open
Abstract
BackgroundMethotrexate (MTX) is the first line treatment for rheumatoid arthritis (RA), but failure of satisfying treatment response occurs in a significant proportion of patients. Here we present a longitudinal multi-omics study aimed at detecting molecular and cellular processes in peripheral blood associated with a successful methotrexate treatment of rheumatoid arthritis.MethodsEighty newly diagnosed patients with RA underwent clinical assessment and donated blood before initiation of MTX, and 3 months into treatment. Flow cytometry was used to describe cell types and presence of activation markers in peripheral blood, the expression of 51 proteins was measured in serum or plasma, and RNA sequencing was performed in peripheral blood mononuclear cells (PBMC). Response to treatment after 3 months was determined using the EULAR response criteria. We assessed the changes in biological phenotypes during treatment, and whether these changes differed between responders and non-responders with regression analysis. By using measurements from baseline, we also tried to find biomarkers of future MTX response or, alternatively, to predict MTX response.ResultsAmong the MTX responders, (Good or Moderate according to EULAR treatment response classification, n = 60, 75%), we observed changes in 29 partly overlapping cell types proportions, levels of 13 proteins and expression of 38 genes during treatment. These changes were in most cases suppressions that were stronger among responders compared to non-responders. Within responders to treatment, we observed a suppression of FOXP3 gene expression, reduction of immunoglobulin gene expression and suppression of genes involved in cell proliferation. The proportion of many HLA-DR expressing T-cell populations were suppressed in all patients irrespective of clinical response, and the proportion of many IL21R+ T-cells were reduced exclusively in non-responders. Using only the baseline measurements we could not detect any biomarkers or prediction models that could predict response to MTX.ConclusionWe conclude that a deep molecular and cellular phenotyping of peripheral blood cells in RA patients treated with methotrexate can reveal previously not recognized differences between responders and non-responders during 3 months of treatment with MTX. This may contribute to the understanding of MTX mode of action and explain non-responsiveness to MTX therapy.
Collapse
Affiliation(s)
- Boel Brynedal
- Translational Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Centre for Epidemiology and Community Medicine, Region Stockholm, Stockholm, Sweden
- *Correspondence: Boel Brynedal,
| | - Niyaz Yoosuf
- Translational Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tinna Bjorg Ulfarsdottir
- Translational Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | - Helga Westerlind
- Translational Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Malin Müller
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peter Sahlström
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Aase Hensvold
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Center for Rheumatology, Academic Specialist Center, Region Stockholm, Stockholm, Sweden
| | - Leonid Padyukov
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Leonid Padyukov,
| | - Nancy Vivar Pomiano
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anca Catrina
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars Klareskog
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Louise Berg
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
Zeng Z, Maiti A, Herbrich S, Cai T, Cavazos A, Manzella T, Ma H, Hayes K, Matthews J, DiNardo CD, Daver NG, Konopleva MY. Triple combination targeting methyltransferase, BCL-2, and PD-1 facilitates antileukemia responses in acute myeloid leukemia. Cancer 2023; 129:531-540. [PMID: 36477735 DOI: 10.1002/cncr.34566] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/04/2022] [Accepted: 09/23/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND A recent breakthrough therapy combining the BCL-2 inhibitor venetoclax with hypomethylating agents (HMAs) targeting DNA methyltransferase has improved outcomes for patients with acute myeloid leukemia (AML), but the responses and long-term survival in older/unfit patients and in patients with relapsed/refractory AML remain suboptimal. Recent studies showed that inhibition of BCL-2 or DNA methyltransferase modulates AML T-cell immunity. METHODS By using flow cytometry and time-of-flight mass cytometry, the authors examined the effects of the HMA decitabine combined with the BCL-2 inhibitor venetoclax (DAC/VEN therapy) on leukemia cells and T cells in patients with AML who received DAC/VEN therapy in a clinical trial. The authors investigated the response of programmed cell death protein 1 (PD-1) inhibition in the DAC/VEN-treated samples in vitro and investigated the triple combination of PD-1 inhibition with HMA/venetoclax in the trial patients who had AML. RESULTS DAC/VEN therapy effectively targeted leukemia cells and upregulated the expression of the immune checkpoint-inhibitory receptor PD-1 in T cells while preserving CD4-positive and CD8-positive memory T cells in a subset of patients with AML who were tested. In vitro PD-1 inhibition potentiated the antileukemia response in DAC/VEN-treated AML samples. The combined use of azacitidine, venetoclax, and nivolumab eliminated circulating blasts and leukemia stem cells/progenitor cells and expanded the percentage of CD8-positive memory T cells in an illustrative patient with relapsed AML who responded to the regimen in an ongoing clinical trial. CONCLUSIONS Immunomodulation by targeting PD-1 enhances the therapeutic effect of combining an HMA and venetoclax in patients with AML.
Collapse
Affiliation(s)
- Zhihong Zeng
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Abhishek Maiti
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shelley Herbrich
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tianyu Cai
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Antonio Cavazos
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Taylor Manzella
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Helen Ma
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kala Hayes
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jairo Matthews
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Courtney D DiNardo
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Naval G Daver
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marina Y Konopleva
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
26
|
Tandel N, Negi S, Dalai SK, Tyagi RK. Role of natural killer and B cell interaction in inducing pathogen specific immune responses. Int Rev Immunol 2023:1-19. [PMID: 36731424 DOI: 10.1080/08830185.2023.2172406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The innate lymphoid cell (ILC) system comprising of the circulating and tissue-resident cells is known to clear infectious pathogens, establish immune homeostasis as well as confer antitumor immunity. Human natural killer cells (hNKs) and other ILCs carry out mopping of the infectious pathogens and perform cytolytic activity regulated by the non-adaptive immune system. The NK cells generate immunological memory and rapid recall response tightly regulated by the adaptive immunity. The interaction of NK and B cell, and its role to induce the pathogen specific immunity is not fully understood. Hence, present article sheds light on the interaction between NK and B cells and resulting immune responses in the infectious diseases. The immune responses elicited by the NK-B cell interaction is of particular importance for developing therapeutic vaccines against the infectious pathogens. Further, experimental evidences suggest the immune-response driven by NK cell population elicits the host-specific antibodies and memory B cells. Also, recently developed humanized immune system (HIS) mice and their importance in to understanding the NK-B cell interaction and resulting pathogen specific immunity has been discussed.
Collapse
Affiliation(s)
- Nikunj Tandel
- Institute of Science, Nirma University, Ahmedabad, India
| | - Sushmita Negi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Nano-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh, India
| | - Sarat K Dalai
- Institute of Science, Nirma University, Ahmedabad, India
| | - Rajeev K Tyagi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Nano-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh, India
| |
Collapse
|
27
|
Choi H, Kim Y, Jung YW. The Function of Memory CD8+ T Cells in Immunotherapy for Human Diseases. Immune Netw 2023; 23:e10. [PMID: 36911798 PMCID: PMC9995995 DOI: 10.4110/in.2023.23.e10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 03/07/2023] Open
Abstract
Memory T (Tm) cells protect against Ags that they have previously contacted with a fast and robust response. Therefore, developing long-lived Tm cells is a prime goal for many vaccines and therapies to treat human diseases. The remarkable characteristics of Tm cells have led scientists and clinicians to devise methods to make Tm cells more useful. Recently, Tm cells have been highlighted for their role in coronavirus disease 2019 vaccines during the ongoing global pandemic. The importance of Tm cells in cancer has been emerging. However, the precise characteristics and functions of Tm cells in these diseases are not completely understood. In this review, we summarize the known characteristics of Tm cells and their implications in the development of vaccines and immunotherapies for human diseases. In addition, we propose to exploit the beneficial characteristics of Tm cells to develop strategies for effective vaccines and overcome the obstacles of immunotherapy.
Collapse
Affiliation(s)
- Hanbyeul Choi
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Yeaji Kim
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Yong Woo Jung
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| |
Collapse
|
28
|
Lu M, Lee Y, Lillehoj HS. Evolution of developmental and comparative immunology in poultry: The regulators and the regulated. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 138:104525. [PMID: 36058383 DOI: 10.1016/j.dci.2022.104525] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 06/15/2023]
Abstract
Avian has a unique immune system that evolved in response to environmental pressures in all aspects of innate and adaptive immune responses, including localized and circulating lymphocytes, diversity of immunoglobulin repertoire, and various cytokines and chemokines. All of these attributes make birds an indispensable vertebrate model for studying the fundamental immunological concepts and comparative immunology. However, research on the immune system in birds lags far behind that of humans, mice, and other agricultural animal species, and limited immune tools have hindered the adequate application of birds as disease models for mammalian systems. An in-depth understanding of the avian immune system relies on the detailed studies of various regulated and regulatory mediators, such as cell surface antigens, cytokines, and chemokines. Here, we review current knowledge centered on the roles of avian cell surface antigens, cytokines, chemokines, and beyond. Moreover, we provide an update on recent progress in this rapidly developing field of study with respect to the availability of immune reagents that will facilitate the study of regulatory and regulated components of poultry immunity. The new information on avian immunity and available immune tools will benefit avian researchers and evolutionary biologists in conducting fundamental and applied research.
Collapse
Affiliation(s)
- Mingmin Lu
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| | - Youngsub Lee
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| |
Collapse
|
29
|
Kuehn J, Schleifenbaum S, Hendling M, Siebenhandl S, Krainer J, Fuehner S, Hellige A, Park C, Hinze C, Wittkowski H, Holzinger D, Thurner L, Weinhäusel A, Foell D, Kessel C. Aberrant Naive CD4-Positive T Cell Differentiation in Systemic Juvenile Idiopathic Arthritis Committed to B Cell Help. Arthritis Rheumatol 2022; 75:826-841. [PMID: 36409585 DOI: 10.1002/art.42409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 11/03/2022] [Accepted: 11/15/2022] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Systemic juvenile idiopathic arthritis (JIA) features characteristics of autoinflammation and autoimmunity, culminating in chronic arthritis. In this study, we hypothesized that aberrant or incomplete polarization of T helper cells contributes to disease pathology. METHODS Cells or serum samples were obtained from healthy controls (n = 72) and systemic JIA patients (n = 171). Isolated naive T helper cells were cultured under Th1, Th17, and T follicular helper (Tfh) or T peripheral helper (Tph)-polarizing conditions and were partly cocultured with allogenic memory B cells. Cell samples were then analyzed for surface marker, transcription factor, and cytokine expression, as well as plasmablast generation. Serum samples were subjected to multiplexed bead and self-antigen arrays and enzyme-linked immunosorbent assays, and all data were compared to retrospective RNA profiling analyses. RESULTS Differentiation of systemic JIA-naive T helper cells toward Th1 cells resulted in low expression levels of interferon-γ (IFNγ) and eomesodermin, which was associated in part with disease duration. In contrast developing Th1 cells in patients with systemic JIA were found to produce elevated levels of interleukin-21 (IL-21), which negatively correlated with cellular expression of IFNγ and eomesodermin. In both in vitro and ex vivo analyses, IL-21 together with programmed cell death 1 (PD-1), inducible T cell costimulator (ICOS), and CXCR5 expression induced naive T helper cells from systemic JIA patients to polarize toward a Tfh/Tph cell phenotype. Retrospective analysis of whole-blood RNA-sequencing data demonstrated that Bcl-6, a master transcription factor in Tfh/Tph cell differentiation, was overexpressed specifically in patients with systemic JIA. Naive T helper cells from systemic JIA patients which were stimulated in vitro promoted B cellular plasmablast generation, and self-antigen array data indicated that IgG reactivity profiles of patients with systemic JIA differed from those of healthy controls. CONCLUSION In the pathogenesis of systemic JIA, skewing of naive T helper cell differentiation toward a Tfh/Tph cell phenotype may represent an echo of autoimmunity, which may indicate the mechanisms driving progression toward chronic destructive arthritis.
Collapse
Affiliation(s)
- Julia Kuehn
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Susanne Schleifenbaum
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Michaela Hendling
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, Austrian Institute of Technology, Vienna, Austria
| | - Sandra Siebenhandl
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, Austrian Institute of Technology, Vienna, Austria
| | - Julie Krainer
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, Austrian Institute of Technology, Vienna, Austria
| | - Sabrina Fuehner
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Antje Hellige
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Carolin Park
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Claas Hinze
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Helmut Wittkowski
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Dirk Holzinger
- Department of Pediatric Hematology-Oncology, University of Duisburg-Essen, Essen, Germany, and Department of Applied Health Sciences, University of Applied Sciences Bochum, Bochum, Germany
| | - Lorenz Thurner
- José Carreras Center for Immunology and Gene Therapy and Internal Medicine I, Saarland University Medical School, Homburg, Germany
| | - Andreas Weinhäusel
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, Austrian Institute of Technology, Vienna, Austria
| | - Dirk Foell
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Christoph Kessel
- Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| |
Collapse
|
30
|
Wang Y, Tian Q, Ye L. The Differentiation and Maintenance of SARS-CoV-2-Specific Follicular Helper T Cells. Front Cell Infect Microbiol 2022; 12:953022. [PMID: 35909969 PMCID: PMC9329515 DOI: 10.3389/fcimb.2022.953022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/20/2022] [Indexed: 12/24/2022] Open
Abstract
Upon acute viral infection, virus-specific CD4+ T cells differentiate into either TH1 cells or follicular helper T (TFH) cells. The molecular pathways governing such bimodal cell fate commitment remain elusive. Additionally, effector virus-specific TFH cells further differentiate into corresponding memory population, which confer long-term protection against re-infection of same viruses by providing immediate help to virus-specific memory B cells. Currently, the molecular mechanisms underlying the long-term maintenance of memory TFH cells are largely unknown. In this review, we discuss current understanding of early differentiation of virus-specific effector TFH cells and long-term maintenance of virus-specific memory TFH cells in mouse models of viral infection and patients of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection.
Collapse
Affiliation(s)
- Yifei Wang
- Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Qin Tian
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- Institute of Immunology, The People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
| | - Lilin Ye
- Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Institute of Immunology, The People’s Liberation Army (PLA), Third Military Medical University, Chongqing, China
- *Correspondence: Lilin Ye,
| |
Collapse
|
31
|
Donlon NE, Davern M, O’Connell F, Sheppard A, Heeran A, Bhardwaj A, Butler C, Narayanasamy R, Donohoe C, Phelan JJ, Lynam-Lennon N, Dunne MR, Maher S, O’Sullivan J, Reynolds JV, Lysaght J. Impact of radiotherapy on the immune landscape in oesophageal adenocarcinoma. World J Gastroenterol 2022; 28:2302-2319. [PMID: 35800186 PMCID: PMC9185220 DOI: 10.3748/wjg.v28.i21.2302] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/19/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND In the contemporary era of cancer immunotherapy, an abundance of clinical and translational studies have reported radiotherapy (RT) and immunotherapies as a viable option for immunomodulation of many cancer subtypes, with many related clinical trials ongoing. In locally advanced disease, chemotherapy or chemoradiotherapy followed by surgical excision of the tumour remain the principal treatment strategy in oesophageal adenocarcinoma (OAC), however, the use of the host immune system to improve anti-tumour immunity is rapidly garnering increased support in the curative setting.
AIM To immunophenotype OAC patients’ immune checkpoint (IC) expression with and without radiation and evaluate the effects of checkpoint blockade on cell viability.
METHODS In the contemporary era of cancer immunotherapy, an abundance of studies have demonstrated that combination RT and IC inhibitors (ICIs) are effective in the immunomodulation of many cancer subtypes, with many related clinical trials ongoing. Although surgical excision and elimination of tumour cells by chemotherapy or chemoradiotherapy remains the gold standard approach in OAC, the propagation of anti-tumour immune responses is rapidly garnering increased support in the curative setting. The aim of this body of work was to immunophenotype OAC patients’ IC expression with and without radiation and to establish the impact of checkpoint blockade on cell viability. This study was a hybrid combination of in vitro and ex vivo models. Quantification of serum immune proteins was performed by enzyme-linked immunosorbent assay. Flow cytometry staining was performed to evaluate IC expression for in vitro OAC cell lines and ex vivo OAC biopsies. Cell viability in the presence of radiation with and without IC blockade was assessed by a cell counting kit-8 assay.
RESULTS We identified that conventional dosing and hypofractionated approaches resulted in increased IC expression (PD-1, PD-L1, TIM3, TIGIT) in vitro and ex vivo in OAC. There were two distinct subcohorts with one demonstrating significant upregulation of ICs and the contrary in the other cohort. Increasing IC expression post RT was associated with a more aggressive tumour phenotype and adverse features of tumour biology. The use of anti-PD-1 and anti-PD-L1 immunotherapies in combination with radiation resulted in a significant and synergistic reduction in viability of both radiosensitive and radioresistant OAC cells in vitro. Interleukin-21 (IL-21) and IL-31 significantly increased, with a concomitant reduction in IL-23 as a consequence of 4 Gray radiation. Similarly, radiation induced an anti-angiogenic tumour milieu with reduced expression of vascular endothelial growth factor-A, basic fibroblast growth factor, Flt-1 and placental growth factor.
CONCLUSION The findings of the current study demonstrate synergistic potential for the use of ICIs and ionising radiation to potentiate established anti-tumour responses in the neoadjuvant setting and is of particular interest in those with advanced disease, adverse features of tumour biology and poor treatment responses to conventional therapies.
Collapse
Affiliation(s)
- Noel E Donlon
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - Maria Davern
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - Fiona O’Connell
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - Andrew Sheppard
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - Aisling Heeran
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - Anshul Bhardwaj
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - Christine Butler
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - Ravi Narayanasamy
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - Claire Donohoe
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - James J Phelan
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - Niamh Lynam-Lennon
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - Margaret R Dunne
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - Stephen Maher
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - Jacintha O’Sullivan
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - John V Reynolds
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| | - Joanne Lysaght
- Department of Surgery, Trinity Translational Medicine Institute, St James Hospital, Dublin D08, Ireland
| |
Collapse
|
32
|
Li D, Cui Z, Zhao F, Zhu X, Tan A, Deng Y, Lai Y, Huang Z. Characterization of snakehead (Channa argus) interleukin-21: Involvement in immune defense against two pathogenic bacteria, in leukocyte proliferation, and in activation of JAK-STAT signaling pathway. FISH & SHELLFISH IMMUNOLOGY 2022; 123:207-217. [PMID: 35278639 DOI: 10.1016/j.fsi.2022.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 06/14/2023]
Abstract
Interleukin-21 (IL-21), a crucial immune regulatory molecule, belongs to the common γ-chain family of type I cytokines, and exerts pleiotropic effects on multiple immune cell types in mammals. However, the characteristics and functions of fish IL-21 remain unclear. To further investigate the molecular mechanism of IL-21 in teleosts, we first cloned and identified the IL-21 gene (designated shIL-21) of the snakehead (Channa argus). The full-length open reading frame of shIL-21 is 438 bp in length, and encodes a predicted protein of 145 amino acid residues. A sequence analysis showed that shIL-21 has the typical structural characteristics of other IL-21 proteins, containing four α-helices and four conserved cysteine residues. In a phylogenetic analysis, shIL-21 clustered within a subgroup of IL-21 proteins from other teleost species and shared its closest evolutionary relationship with that of Lates calcarifer. The expression analysis showed that shIL-21 was ubiquitously expressed in all the healthy snakehead tissues tested, albeit at different levels. After infection with Nocardia seriolae or Aeromonas schubertii, the relative expression of shIL-21 was mainly upregulated in the head kidney and spleen in vivo. Similarly, after stimulation with the three pathogen analogues lipoteichoic acid, lipopolysaccharides, and polyinosinic-polycytidylic acid, the expression of shIL-21 was also induced in head kidney leukocytes in vitro. A recombinant shIL-21 protein was expressed and purified, and promoted the proliferation of head kidney leukocytes, induced the expression of genes encoding critical signaling molecules in the Janus kinase (JAK) and signal transducer and activator of transcription (STAT) pathway, including JAK1, JAK3, STAT1, and STAT3, and induced the expression of endogenous shIL-21 and genes encoding several key proinflammatory cytokines (tumor necrosis factor-α, interferon-γ, and IL-1β). Taken together, these preliminary findings suggest that shIL-21 is involved in the immune defense against bacterial infection, in leukocyte proliferation, and in the activation of the JAK-STAT pathway. They thus extend the functional studies of IL-21 in teleosts.
Collapse
Affiliation(s)
- Dongqi Li
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Zhengwei Cui
- Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Fei Zhao
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China.
| | - Xueqing Zhu
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Aiping Tan
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Yuting Deng
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Yingtiao Lai
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Zhibin Huang
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| |
Collapse
|
33
|
Holder PG, Lim SA, Huang CS, Sharma P, Dagdas YS, Bulutoglu B, Sockolosky JT. Engineering interferons and interleukins for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114112. [PMID: 35085624 DOI: 10.1016/j.addr.2022.114112] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Cytokines are a class of potent immunoregulatory proteins that are secreted in response to various stimuli and act locally to regulate many aspects of human physiology and disease. Cytokines play important roles in cancer initiation, progression, and elimination, and thus, there is a long clinical history associated with the use of recombinant cytokines to treat cancer. However, the use of cytokines as therapeutics has been limited by cytokine pleiotropy, complex biology, poor drug-like properties, and severe dose-limiting toxicities. Nevertheless, cytokines are crucial mediators of innate and adaptive antitumor immunity and have the potential to enhance immunotherapeutic approaches to treat cancer. Development of immune checkpoint inhibitors and combination immunotherapies has reinvigorated interest in cytokines as therapeutics, and a variety of engineering approaches are emerging to improve the safety and effectiveness of cytokine immunotherapy. In this review we highlight recent advances in cytokine biology and engineering for cancer immunotherapy.
Collapse
|
34
|
Beyond immune checkpoint blockade: emerging immunological strategies. Nat Rev Drug Discov 2021; 20:899-919. [PMID: 33686237 DOI: 10.1038/s41573-021-00155-y] [Citation(s) in RCA: 235] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2021] [Indexed: 02/07/2023]
Abstract
The success of checkpoint inhibitors has accelerated the clinical implementation of a vast mosaic of single agents and combination immunotherapies. However, the lack of clinical translation for a number of immunotherapies as monotherapies or in combination with checkpoint inhibitors has clarified that new strategies must be employed to advance the field. The next chapter of immunotherapy should examine the immuno-oncology therapeutic failures, and consider the complexity of immune cell-cancer cell interactions to better design more effective anticancer drugs. Herein, we briefly review the history of immunotherapy and checkpoint blockade, highlighting important clinical failures. We discuss the critical aspects - beyond T cell co-receptors - of immune processes within the tumour microenvironment (TME) that may serve as avenues along which new therapeutic strategies in immuno-oncology can be forged. Emerging insights into tumour biology suggest that successful future therapeutics will focus on two key factors: rescuing T cell homing and dysfunction in the TME, and reappropriating mononuclear phagocyte function for TME inflammatory remodelling. New drugs will need to consider the complex cell networks that exist within tumours and among cancer types.
Collapse
|
35
|
Kim DH, Kim HY, Lee WW. Induction of Unique STAT Heterodimers by IL-21 Provokes IL-1RI Expression on CD8 + T Cells, Resulting in Enhanced IL-1β Dependent Effector Function. Immune Netw 2021; 21:e33. [PMID: 34796037 PMCID: PMC8568912 DOI: 10.4110/in.2021.21.e33] [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: 05/25/2021] [Revised: 08/11/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022] Open
Abstract
IL-1β plays critical roles in the priming and effector phases of immune responses such as the differentiation, commitment, and memory formation of T cells. In this context, several reports have suggested that the IL-1β signal is crucial for CTL-mediated immune responses to viral infections and tumors. However, little is known regarding whether IL-1β acts directly on CD8+ T cells and what the molecular mechanisms underlying expression of IL-1 receptors (IL-1Rs) on CD8+ T cells and features of IL-1R+CD8+ T cells are. Here, we provide evidence that the expression of IL-1R type I (IL-1RI), the functional receptor of IL-1β, is preferentially induced by IL-21 on TCR-stimulated CD8+ T cells. Further, IL-1β enhances the effector function of CD8+ T cells expressing IL-21-induced IL-1RI by increasing cytokine production and release of cytotoxic granules containing granzyme B. The IL-21-IL-1RI-IL-1β axis is involved in an augmented effector function through regulation of transcription factors BATF, Blimp-1, and IRF4. Moreover, this axis confers a unique effector function to CD8+ T cells compared to conventional type 1 cytotoxic T cells differentiated with IL-12. Chemical inhibitor and immunoprecipitation assay demonstrated that IL-21 induces a unique pattern of STAT activation with the formation of both STAT1:STAT3 and STAT3:STAT5 heterodimers, which are critical for the induction of IL-1RI on TCR-stimulated CD8+ T cells. Taken together, we propose that induction of a novel subset of IL-1RI-expressing CD8+ T cells by IL-21 may be beneficial to the protective immune response against viral infections and is therefore important to consider for vaccine design.
Collapse
Affiliation(s)
- Dong Hyun Kim
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hee Young Kim
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea.,Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Won-Woo Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea.,Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul 03080, Korea.,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea.,Seoul National University Hospital Biomedical Research Institute, Seoul 03080, Korea
| |
Collapse
|
36
|
Peng YM, Tao JJ, Kuang SF, Jiang M, Peng XX, Li H. Identification of Polyvalent Vaccine Candidates From Extracellular Secretory Proteins in Vibrio alginolyticus. Front Immunol 2021; 12:736360. [PMID: 34671354 PMCID: PMC8521057 DOI: 10.3389/fimmu.2021.736360] [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: 07/05/2021] [Accepted: 09/14/2021] [Indexed: 12/18/2022] Open
Abstract
Bacterial infections cause huge losses in aquaculture and a wide range of health issues in humans. A vaccine is the most economical, efficient, and environment-friendly agent for protecting hosts against bacterial infections. This study aimed to identify broad, cross-protective antigens from the extracellular secretory proteome of the marine bacterium Vibrio alginolyticus. Of the 69 predicted extracellular secretory proteins in its genome, 16 were randomly selected for gene cloning to construct DNA vaccines, which were used to immunize zebrafish (Danio rerio). The innate immune response genes were also investigated. Among the 16 DNA vaccines, 3 (AT730_21605, AT730_22220, and AT730_22910) were protective against V. alginolyticus infection with 47–66.7% increased survival compared to the control, while other vaccines had lower or no protective effects. Furthermore, AT730_22220, AT730_22910, and AT730_21605 also exhibited cross-immune protective effects against Pseudomonas fluorescens and/or Aeromonas hydrophila infection. Mechanisms for cross-protective ability was explored based on conserved epitopes, innate immune responses, and antibody neutralizing ability. These results indicate that AT730_21605, AT730_22220, and AT730_22910 are potential polyvalent vaccine candidates against bacterial infections. Additionally, our results suggest that the extracellular secretory proteome is an antigen pool that can be used for the identification of cross-protective immunogens.
Collapse
Affiliation(s)
- Yu-Ming Peng
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
| | - Jian-Jun Tao
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
| | - Su-Fang Kuang
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
| | - Ming Jiang
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
| | - Xuan-Xian Peng
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Hui Li
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
37
|
Czarnowicki T, Kim HJ, Villani AP, Glickman J, Duca ED, Han J, Pavel AB, Lee BH, Rahman AH, Merad M, Krueger JG, Guttman‐Yassky E. High-dimensional analysis defines multicytokine T-cell subsets and supports a role for IL-21 in atopic dermatitis. Allergy 2021; 76:3080-3093. [PMID: 33818809 DOI: 10.1111/all.14845] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/12/2021] [Accepted: 01/17/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Flow cytometry is a well-accepted approach for immune profiling; however, its value is restricted by the limited number of markers that can be analyzed simultaneously. Mass cytometry/CyTOF offers broad-scale immune characterization integrating large number of parameters. While partial blood phenotyping was reported in atopic dermatitis (AD), patients' comprehensive profiling, critical for leveraging new targeted treatments, is not available. IL-21 may be involved in inflammatory skin diseases but its role in AD is not well established. METHODS We studied T-cell polarization in the blood of 20 moderate-to-severe AD and 15 controls. Using CyTOF and an unsupervised analysis, we measured the frequencies and mean metal intensities of activated polar CD4+ /CD8+ T-cell subsets. Immunohistochemistry, immunofluorescence, and qRT-PCR were used to analyze skin samples. RESULTS Examining 24 surface, intracellular markers, and transcription factors, we identified six CD4+ and five CD8+ T-cell metaclusters. A CD4+ skin-homing IL-13+ monocytokine and a novel IL-13+ IL-21+ multicytokine metaclusters were increased in AD vs. controls (p < .01). While IL-13 signature characterized both clusters, levels were significantly higher in the IL-21+ group. Both clusters correlated with AD severity (r = 0.49, p = .029). Manual gating corroborated these results and identified additional multicytokine subsets in AD. Immunohistochemistry and immunofluorescence, validated by mRNA expression, displayed significantly increasedIL-21 counts and colocalization with IL-13/IL-4R in AD skin. CONCLUSION A multicytokine signature characterizes moderate-to-severe AD, possibly explaining partial therapeutic responses to one cytokine targeting, particularly in severe patients. Prominent IL-21 signature in blood and skin hints for a potential pathogenic role of IL-21 in AD.
Collapse
Affiliation(s)
- Tali Czarnowicki
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
- Laboratory for Investigative Dermatology The Rockefeller University New York NY USA
| | - Hyun Je Kim
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Axel P. Villani
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Jacob Glickman
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Ester Del Duca
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Joseph Han
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Ana B. Pavel
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| | - Brian H. Lee
- Human Immune Monitoring Center Icahn School of Medicine at Mt. Sinai New York NY USA
| | - Adeeb H. Rahman
- Human Immune Monitoring Center Icahn School of Medicine at Mt. Sinai New York NY USA
- Department of Genetics and Genomic Sciences Icahn School of Medicine at Mount Sinai New York NY USA
| | - Miriam Merad
- Department of Oncological Sciences Icahn School of Medicine at Mount Sinai New York NY USA
- Icahn School of Medicine at Mount Sinai The Precision Immunology Institute New York NY USA
- Icahn School of Medicine at Mount Sinai The Tisch Cancer Institute New York NY USA
| | - James G. Krueger
- Laboratory for Investigative Dermatology The Rockefeller University New York NY USA
| | - Emma Guttman‐Yassky
- Department of Dermatology and the Immunology Institute Icahn School of Medicine at Mount Sinai New York NY USA
| |
Collapse
|
38
|
Ma X, Somasundaram A, Qi Z, Hartman D, Singh H, Osmanbeyoglu H. SPaRTAN, a computational framework for linking cell-surface receptors to transcriptional regulators. Nucleic Acids Res 2021; 49:9633-9647. [PMID: 34500467 PMCID: PMC8464045 DOI: 10.1093/nar/gkab745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/09/2021] [Accepted: 09/06/2021] [Indexed: 12/22/2022] Open
Abstract
The identity and functions of specialized cell types are dependent on the complex interplay between signaling and transcriptional networks. Recently single-cell technologies have been developed that enable simultaneous quantitative analysis of cell-surface receptor expression with transcriptional states. To date, these datasets have not been used to systematically develop cell-context-specific maps of the interface between signaling and transcriptional regulators orchestrating cellular identity and function. We present SPaRTAN (Single-cell Proteomic and RNA based Transcription factor Activity Network), a computational method to link cell-surface receptors to transcription factors (TFs) by exploiting cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) datasets with cis-regulatory information. SPaRTAN is applied to immune cell types in the blood to predict the coupling of signaling receptors with cell context-specific TFs. Selected predictions are validated by prior knowledge and flow cytometry analyses. SPaRTAN is then used to predict the signaling coupled TF states of tumor infiltrating CD8+ T cells in malignant peritoneal and pleural mesotheliomas. SPaRTAN enhances the utility of CITE-seq datasets to uncover TF and cell-surface receptor relationships in diverse cellular states.
Collapse
Affiliation(s)
- Xiaojun Ma
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Ashwin Somasundaram
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Zengbiao Qi
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Douglas J Hartman
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Harinder Singh
- Center for Systems Immunology and Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Hatice Ulku Osmanbeyoglu
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
39
|
Xu T, Pereira RM, Martinez GJ. An Updated Model for the Epigenetic Regulation of Effector and Memory CD8 + T Cell Differentiation. THE JOURNAL OF IMMUNOLOGY 2021; 207:1497-1505. [PMID: 34493604 DOI: 10.4049/jimmunol.2100633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 07/22/2021] [Indexed: 11/19/2022]
Abstract
Naive CD8+ T cells, upon encountering their cognate Ag in vivo, clonally expand and differentiate into distinct cell fates, regulated by transcription factors and epigenetic modulators. Several models have been proposed to explain the differentiation of CTLs, although none fully recapitulate the experimental evidence. In this review article, we will summarize the latest research on the epigenetic regulation of CTL differentiation as well as provide a combined model that contemplates them.
Collapse
Affiliation(s)
- Tianhao Xu
- Discipline of Microbiology and Immunology, Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL; and
| | - Renata M Pereira
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gustavo J Martinez
- Discipline of Microbiology and Immunology, Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL; and
| |
Collapse
|
40
|
Xie L, Zhang Z, Zhu P, Tian K, Liu Y, Yu Y. IL-21 Prevents Expansion of CD8 +CD28 - T Cells Stimulated by IL-15 and Changes Their Subset Distribution. Transplant Proc 2021; 53:2407-2414. [PMID: 34474914 DOI: 10.1016/j.transproceed.2021.07.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/20/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND To examine the effect of interleukin (IL)-21 on the proliferation, subsets, and immunological characteristics of CD8+CD28- T cells stimulated by IL-15 in vitro. METHODS Purified CD8+ T cells stimulated with allogeneic CD2- cells obtained from the peripheral blood mononuclear cells of healthy volunteers were cocultured in the presence of IL-15 alone or IL-21 and IL-15 combined. The dynamic changes in the proliferation, subsets, and phenotypic characteristics of CD8+CD28- T cells were detected. Our work, involving human participants, complied with the Declaration of Helsinki and the Declaration of Istanbul. RESULTS IL-21 prevented the expansion of CD8+CD28- T cells stimulated by IL-15 by sustaining CD28 expression at the mRNA level. IL-15 altered the expanded CD8+CD28- T cell memory subsets over the coculture duration, but the addition of IL-21 could change the subset distribution. In the presence of IL-15, the in vitro-expanded CD8+CD28- T cells were mainly intermediately differentiated cells, but they were mainly late differentiated cells in the presence of IL-21 plus IL-15. Moreover, IL-21 upregulated the expression of toxic molecules in the IL-15-expanded CD8+CD28- T cells. CONCLUSIONS IL-21 prevents IL-15-induced CD8+CD28- T cell amplification by downregulating CD28 at the transcriptional level. IL-21 can alter the subpopulation distribution and phenotypic characteristics of CD8+CD28- T cells stimulated by IL-15.
Collapse
Affiliation(s)
- Lu Xie
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Zedan Zhang
- Shantou University Medical College, Shantou, Guangdong, China
| | - Ping Zhu
- Department of Immunology, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, China
| | - Kaiwen Tian
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yanjun Liu
- Department of Immunology, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuming Yu
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
| |
Collapse
|
41
|
Bayraktar N, Turan H, Bayraktar M, Ozturk A, Erdoğdu H. Analysis of serum cytokine and protective vitamin D levels in severe cases of COVID-19. J Med Virol 2021; 94:154-160. [PMID: 34427934 PMCID: PMC8661791 DOI: 10.1002/jmv.27294] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/06/2021] [Accepted: 08/14/2021] [Indexed: 12/24/2022]
Abstract
In this study, we investigated the role and relationship between the cytokine profile and protective vitamin D by measuring their serum levels in COVID‐19 intensive care unit patients with severe illnesses. A total of 74 patients were included in our study. Patients were divided into two groups. Patients in the COVID‐19 group (n = 31) and individuals without a history of serious illness or infection were used as the control group (n = 43). The serum concentrations of interleukin‐1 (IL‐1), IL‐6, IL‐10, IL‐21, and tumor necrosis factor‐α (TNF‐α) were measured by enzyme‐linked immunosorbent assays. Levels of serum vitamin D were detected with Liquid chromatography–mass spectrometry methodologies. TNF‐α, IL‐1, IL‐6, IL‐10, IL‐21, and vitamin D levels were measured in all patients. The serum cytokine levels in the COVID‐19 patient group were significantly higher (151.59 ± 56.50, 140.37 ± 64.32, 249.02 ± 62.84, 129.04 ± 31.64, and 123.58 ± 24.49, respectively) than control groups. Serum vitamin D was also significantly low (6.82 ± 3.29) in patients in the COVID‐19 group than the controls (21.96 ± 5.39). Regarding the correlation of vitamin D with cytokine levels, it was significantly variable. Our study shows that COVID‐19 patients are associated with lower serum vitamin D and higher pro‐inflammatory cytokines associated with increased virus presence. Our data provide more evidence of the anti‐inflammatory effect of vitamin D on COVID‐19 patients and the protective effects of vitamin D on risk were demonstrated.
Collapse
Affiliation(s)
- Nihayet Bayraktar
- Department of Medical Biochemistry, Faculty of Medicine, Harran University, Şanlıurfa, Turkey
| | - Hamdiye Turan
- Department of Chest Diseases, Faculty of Medicine, Harran University, Şanlıurfa, Turkey
| | - Mehmet Bayraktar
- Department of Medical Microbiology, Faculty of Medicine, Harran University, Şanlıurfa, Turkey
| | - Ali Ozturk
- Department of Medical Microbiology, Faculty of Medicine, Nigde Omer Halisdemir University, Nigde, Turkey
| | - Hamza Erdoğdu
- Department of Statistics, Faculty of Administration and Economics, Harran University, Şanlıurfa, Turkey
| |
Collapse
|
42
|
Zhang Z, Miao L, Ren Z, Tang F, Li Y. Gene-Edited Interleukin CAR-T Cells Therapy in the Treatment of Malignancies: Present and Future. Front Immunol 2021; 12:718686. [PMID: 34386015 PMCID: PMC8353254 DOI: 10.3389/fimmu.2021.718686] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/09/2021] [Indexed: 01/05/2023] Open
Abstract
In recent years, chimeric antigen receptor T cells (CAR-T cells) have been faced with the problems of weak proliferation and poor persistence in the treatment of some malignancies. Researchers have been trying to perfect the function of CAR-T by genetically modifying its structure. In addition to the participation of T cell receptor (TCR) and costimulatory signals, immune cytokines also exert a decisive role in the activation and proliferation of T cells. Therefore, genetic engineering strategies were used to generate cytokines to enhance tumor killing function of CAR-T cells. When CAR-T cells are in contact with target tumor tissue, the proliferation ability and persistence of T cells can be improved by structurally or inductively releasing immunoregulatory molecules to the tumor region. There are a large number of CAR-T cells studies on gene-edited cytokines, and the most common cytokines involved are interleukins (IL-7, IL-12, IL-15, IL-18, IL-21, IL-23). Methods for the construction of gene-edited interleukin CAR-T cells include co-expression of single interleukin, two interleukin, interleukin combined with other cytokines, interleukin receptors, interleukin subunits, and fusion inverted cytokine receptors (ICR). Preclinical and clinical trials have yielded positive results, and many more are under way. By reading a large number of literatures, we summarized the functional characteristics of some members of the interleukin family related to tumor immunotherapy, and described the research status of gene-edited interleukin CAR-T cells in the treatment of malignant tumors. The objective is to explore the optimized strategy of gene edited interleukin-CAR-T cell function.
Collapse
Affiliation(s)
- Zhengchao Zhang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Lele Miao
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Zhijian Ren
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Futian Tang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Yumin Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
43
|
Darden DB, Ghita GL, Wang Z, Stortz JA, Lopez MC, Cox MC, Hawkins RB, Rincon JC, Kelly LS, Fenner BP, Ozrazgat-Baslanti T, Leeuwenburgh C, Bihorac A, Loftus TJ, Moore FA, Brakenridge SC, Baker HV, Bacher R, Mohr AM, Moldawer LL, Efron PA. Chronic Critical Illness Elicits a Unique Circulating Leukocyte Transcriptome in Sepsis Survivors. J Clin Med 2021; 10:3211. [PMID: 34361995 PMCID: PMC8348105 DOI: 10.3390/jcm10153211] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
Surgical sepsis has evolved into two major subpopulations: patients who rapidly recover, and those who develop chronic critical illness (CCI). Our primary aim was to determine whether CCI sepsis survivors manifest unique blood leukocyte transcriptomes in late sepsis that differ from transcriptomes among sepsis survivors with rapid recovery. In a prospective cohort study of surgical ICU patients, genome-wide expression analysis was conducted on total leukocytes in human whole blood collected on days 1 and 14 from sepsis survivors who rapidly recovered or developed CCI, defined as ICU length of stay ≥ 14 days with persistent organ dysfunction. Both sepsis patients who developed CCI and those who rapidly recovered exhibited marked changes in genome-wide expression at day 1 which remained abnormal through day 14. Although summary changes in gene expression were similar between CCI patients and subjects who rapidly recovered, CCI patients exhibited differential expression of 185 unique genes compared with rapid recovery patients at day 14 (p < 0.001). The transcriptomic patterns in sepsis survivors reveal an ongoing immune dyscrasia at the level of the blood leukocyte transcriptome, consistent with persistent inflammation and immune suppression. Furthermore, the findings highlight important genes that could compose a prognostic transcriptomic metric or serve as therapeutic targets among sepsis patients that develop CCI.
Collapse
Affiliation(s)
- Dijoia B. Darden
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Gabriela L. Ghita
- Department of Biostatistics, University of Florida, Gainesville, FL 32610, USA; (G.L.G.); (Z.W.); (R.B.)
| | - Zhongkai Wang
- Department of Biostatistics, University of Florida, Gainesville, FL 32610, USA; (G.L.G.); (Z.W.); (R.B.)
| | - Julie A. Stortz
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Maria-Cecilia Lopez
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA; (M.-C.L.); (H.V.B.)
| | - Michael C. Cox
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Russell B. Hawkins
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Jaimar C. Rincon
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Lauren S. Kelly
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Brittany P. Fenner
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Tezcan Ozrazgat-Baslanti
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL 32610, USA; (T.O.-B.); (C.L.)
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL 32610, USA; (T.O.-B.); (C.L.)
| | - Azra Bihorac
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA;
| | - Tyler J. Loftus
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Frederick A. Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Scott C. Brakenridge
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Henry V. Baker
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA; (M.-C.L.); (H.V.B.)
| | - Rhonda Bacher
- Department of Biostatistics, University of Florida, Gainesville, FL 32610, USA; (G.L.G.); (Z.W.); (R.B.)
| | - Alicia M. Mohr
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Lyle L. Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Philip A. Efron
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| |
Collapse
|
44
|
Ren HM, Lukacher AE, Rahman ZSM, Olsen NJ. New developments implicating IL-21 in autoimmune disease. J Autoimmun 2021; 122:102689. [PMID: 34224936 DOI: 10.1016/j.jaut.2021.102689] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 06/23/2021] [Indexed: 01/01/2023]
Abstract
Elevated interleukin (IL)-21 is a common finding in the tissues and/or sera of patients with autoimmune disease. CD4 T cells are the primary producers of IL-21; often the IL-21 producing CD4 T cells will express molecules associated with follicular helper cells (TFH). Recent work has shown that the CD4 T cell-derived IL-21 is able to promote effector functions and memory differentiation of CD8 T cells in chronic infections and cancer. Autoimmunity has similarities to chronic infections and cancer. However, CD4 T cell-derived IL-21:IL21R signaling in CD8 T cells has not been fully appreciated in the context of autoimmunity. In this review, we assess the current knowledge regarding CD4 T cell-derived IL-21 and IL21R signaling within CD8 T cells and evaluate what implications it has within several autoimmune diseases including systemic lupus erythematous, rheumatoid arthritis, juvenile idiopathic arthritis, type 1 diabetes mellitus, psoriasis, Sjögren's syndrome, vitiligo, antiphospholipid syndrome, pemphigus, and giant cell arteritis.
Collapse
Affiliation(s)
- Heather M Ren
- MD/PhD Medical Scientist Training Program at Penn State College of Medicine, Penn State College of Medicine, Hershey, PA, 17033, USA; Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA, 17033, USA.
| | - Aron E Lukacher
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Ziaur S M Rahman
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Nancy J Olsen
- Devision of Rheumatology, Department of Medicine, Penn State MS Hershey Medical Center, Hershey, PA, 17033, USA
| |
Collapse
|
45
|
The Role of Th17 Response in COVID-19. Cells 2021; 10:cells10061550. [PMID: 34205262 PMCID: PMC8235311 DOI: 10.3390/cells10061550] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/22/2022] Open
Abstract
COVID-19 is an acute infectious disease of the respiratory system caused by infection with the SARS-CoV-2 virus (Severe Acute Respiratory Syndrome Coronavirus 2). Transmission of SARS-CoV-2 infections occurs through droplets and contaminated objects. A rapid and well-coordinated immune system response is the first line of defense in a viral infection. However, a disturbed and over-activated immune response may be counterproductive, causing damage to the body. Severely ill patients hospitalised with COVID-19 exhibit increased levels of many cytokines, including Interleukin (IL)-1β, IL-2, IL-6, IL-7, IL-8, IL-10, IL-17, granulocyte colony stimulating factor (G-CSF), monocyte chemoattractant protein 1 (MCP-1) and tumor necrosis factor (TNF). Increasing evidence suggests that Th17 cells play an important role in the pathogenesis of COVID-19, not only by activating cytokine cascade but also by inducing Th2 responses, inhibiting Th1 differentiation and suppressing Treg cells. This review focuses on a Th17 pathway in the course of the immune response in COVID-19, and explores plausible targets for therapeutic intervention.
Collapse
|
46
|
Zhao Y, Zhang Z, Lei W, Wei Y, Ma R, Wen Y, Wei F, Fan J, Xu Y, Chen L, Lyu K, Lin H, Wen W, Sun W. IL-21 Is an Accomplice of PD-L1 in the Induction of PD-1-Dependent Treg Generation in Head and Neck Cancer. Front Oncol 2021; 11:648293. [PMID: 34026621 PMCID: PMC8131831 DOI: 10.3389/fonc.2021.648293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/19/2021] [Indexed: 11/13/2022] Open
Abstract
Regulatory T cells (Tregs) are immunosuppressive cells involved in antitumor immunity. However, the regulation of Treg generation by inflammation in the tumor microenvironment has not been carefully investigated. Here, we demonstrated that IL-21-polarized inflammation was enriched in the tumor microenvironment in head and neck squamous cell carcinoma (HNSCC) and that IL-21 could promote PD-L1-induced Treg generation in a PD-1-dependent manner. Moreover, generated Tregs showed a greater ability to suppress the proliferation of tumor-associated antigen (TAA)-specific T cells than naturally occurring Tregs. Importantly, an anti-PD-1 antibody could inhibit only Treg expansion induced by clinical tumor explants with high expression of IL-21/PD-L1. In addition, neutralizing IL-21 could enhance the anti-PD-1 antibody-mediated inhibitory effect on Treg expansion. Furthermore, simultaneous high expression of IL-21 and PD-L1 was associated with more Treg infiltrates and predicted reduced overall and disease-free survival in patients with HNSCC. These findings indicate that IL-21 in the tumor microenvironment may promote PD-L1-induced, Treg-mediated immune escape in a PD-1-dependent manner and that an IL-21 neutralization strategy may enhance PD-1 blockade-based antitumor immunotherapy by targeting Treg-mediated immune evasion in patients with high expression of IL-21 and PD-L1.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhiyu Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenbin Lei
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yi Wei
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Renqiang Ma
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yihui Wen
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fanqin Wei
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Fan
- Department of Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, China
| | - Yang Xu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lin Chen
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kexing Lyu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hanqing Lin
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Weiping Wen
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Sun
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
47
|
Niu W, Xu Y, Zha X, Zeng J, Qiao S, Yang S, Zhang H, Tan L, Sun L, Pang G, Liu T, Zhao H, Zheng N, Zhang Y, Bai H. IL-21/IL-21R Signaling Aggravated Respiratory Inflammation Induced by Intracellular Bacteria through Regulation of CD4 + T Cell Subset Responses. THE JOURNAL OF IMMUNOLOGY 2021; 206:1586-1596. [PMID: 33608454 DOI: 10.4049/jimmunol.2001107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/19/2021] [Indexed: 12/16/2022]
Abstract
The IL-21/IL-21R interaction plays an important role in a variety of immune diseases; however, the roles and mechanisms in intracellular bacterial infection are not fully understood. In this study, we explored the effect of IL-21/IL-21R on chlamydial respiratory tract infection using a chlamydial respiratory infection model. The results showed that the mRNA expression of IL-21 and IL-21R was increased in Chlamydia muridarum-infected mice, which suggested that IL-21 and IL-21R were involved in host defense against C. muridarum lung infection. IL-21R-/- mice exhibited less body weight loss, a lower bacterial burden, and milder pathological changes in the lungs than wild-type (WT) mice during C. muridarum lung infection. The absolute number and activity of CD4+ T cells and the strength of Th1/Th17 responses in IL-21R-/- mice were significantly higher than those in WT mice after C. muridarum lung infection, but the Th2 response was weaker. Consistently, IL-21R-/- mice showed higher mRNA expression of Th1 transcription factors (T-bet/STAT4), IL-12p40, a Th17 transcription factor (STAT3), and IL-23. The mRNA expression of Th2 transcription factors (GATA3/STAT6), IL-4, IL-10, and TGF-β in IL-21R-/- mice was significantly lower than that in WT mice. Furthermore, the administration of recombinant mouse IL-21 aggravated chlamydial lung infection in C57BL/6 mice and reduced Th1 and Th17 responses following C. muridarum lung infection. These findings demonstrate that IL-21/IL-21R may aggravate chlamydial lung infection by inhibiting Th1 and Th17 responses.
Collapse
Affiliation(s)
- Wenhao Niu
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Yueyue Xu
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Xiaoyu Zha
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Jiajia Zeng
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Sai Qiao
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Shuaini Yang
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Hong Zhang
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Lu Tan
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Lida Sun
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Gaoju Pang
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Tengli Liu
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Huili Zhao
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Ningbo Zheng
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Yongci Zhang
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Hong Bai
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, People's Republic of China
| |
Collapse
|
48
|
Zhang Y, Li B, Bai Q, Wang P, Wei G, Li Z, Hu L, Tian Q, Zhou J, Huang Q, Wang Z, Yue S, Wu J, Yang L, Zhou X, Jiang L, Ni T, Ye L, Wu Y. The lncRNA Snhg1-Vps13D vesicle trafficking system promotes memory CD8 T cell establishment via regulating the dual effects of IL-7 signaling. Signal Transduct Target Ther 2021; 6:126. [PMID: 33758164 PMCID: PMC7987995 DOI: 10.1038/s41392-021-00492-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/18/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
The efficient induction and long-term persistence of pathogen-specific memory CD8 T cells are pivotal to rapidly curb the reinfection. Recent studies indicated that long-noncoding RNAs expression is highly cell- and stage-specific during T cell development and differentiation, suggesting their potential roles in T cell programs. However, the key lncRNAs playing crucial roles in memory CD8 T cell establishment remain to be clarified. Through CD8 T cell subsets profiling of lncRNAs, this study found a key lncRNA-Snhg1 with the conserved naivehi-effectorlo-memoryhi expression pattern in CD8 T cells of both mice and human, that can promote memory formation while impeding effector CD8 in acute viral infection. Further, Snhg1 was found interacting with the conserved vesicle trafficking protein Vps13D to promote IL-7Rα membrane location specifically. With the deep mechanism probing, the results show Snhg1-Vps13D regulated IL-7 signaling with its dual effects in memory CD8 generation, which not just because of the sustaining role of STAT5-BCL-2 axis for memory survival, but more through the STAT3-TCF1-Blimp1 axis for transcriptional launch program of memory differentiation. Moreover, we performed further study with finding a similar high-low-high expression pattern of human SNHG1/VPS13D/IL7R/TCF7 in CD8 T cell subsets from PBMC samples of the convalescent COVID-19 patients. The central role of Snhg1-Vps13D-IL-7R-TCF1 axis in memory CD8 establishment makes it a potential target for improving the vaccination effects to control the ongoing pandemic.
Collapse
Affiliation(s)
- Yanyan Zhang
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China. .,Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 401121, China.
| | - Baohua Li
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China
| | - Qiang Bai
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China.,Laboratory of Immunophysiology, GIGA Institute, Liège University, Liège, 4000, Belgium.,Faculty of Veterinary Medicine, Liège University, Liège, 4000, Belgium
| | - Pengcheng Wang
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China
| | - Gang Wei
- Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Zhirong Li
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China
| | - Li Hu
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China
| | - Qin Tian
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China
| | - Jing Zhou
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China
| | - Qizhao Huang
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China
| | - Zhiming Wang
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China
| | - Shuai Yue
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China
| | - Jialin Wu
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China
| | - Liuqing Yang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, 77030, TX, USA
| | - Xinyuan Zhou
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China
| | - Lubin Jiang
- Institute Pasteur of Shanghai, Chinese Academy of Sciences (CAS), Shanghai, 200031, China
| | - Ting Ni
- Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Lilin Ye
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China.
| | - Yuzhang Wu
- Institute of Immunology PLA, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
49
|
Zhang Y, Li B, Bai Q, Wang P, Wei G, Li Z, Hu L, Tian Q, Zhou J, Huang Q, Wang Z, Yue S, Wu J, Yang L, Zhou X, Jiang L, Ni T, Ye L, Wu Y. The lncRNA Snhg1-Vps13D vesicle trafficking system promotes memory CD8 T cell establishment via regulating the dual effects of IL-7 signaling. Signal Transduct Target Ther 2021. [DOI: https://doi.org/10.1038/s41392-021-00492-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
AbstractThe efficient induction and long-term persistence of pathogen-specific memory CD8 T cells are pivotal to rapidly curb the reinfection. Recent studies indicated that long-noncoding RNAs expression is highly cell- and stage-specific during T cell development and differentiation, suggesting their potential roles in T cell programs. However, the key lncRNAs playing crucial roles in memory CD8 T cell establishment remain to be clarified. Through CD8 T cell subsets profiling of lncRNAs, this study found a key lncRNA-Snhg1 with the conserved naivehi-effectorlo-memoryhi expression pattern in CD8 T cells of both mice and human, that can promote memory formation while impeding effector CD8 in acute viral infection. Further, Snhg1 was found interacting with the conserved vesicle trafficking protein Vps13D to promote IL-7Rα membrane location specifically. With the deep mechanism probing, the results show Snhg1-Vps13D regulated IL-7 signaling with its dual effects in memory CD8 generation, which not just because of the sustaining role of STAT5-BCL-2 axis for memory survival, but more through the STAT3-TCF1-Blimp1 axis for transcriptional launch program of memory differentiation. Moreover, we performed further study with finding a similar high-low-high expression pattern of human SNHG1/VPS13D/IL7R/TCF7 in CD8 T cell subsets from PBMC samples of the convalescent COVID-19 patients. The central role of Snhg1-Vps13D-IL-7R-TCF1 axis in memory CD8 establishment makes it a potential target for improving the vaccination effects to control the ongoing pandemic.
Collapse
|
50
|
Guo K, Zhang X. Cytokines that Modulate the Differentiation of Th17 Cells in Autoimmune Uveitis. J Immunol Res 2021; 2021:6693542. [PMID: 33816637 PMCID: PMC7990547 DOI: 10.1155/2021/6693542] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/01/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence has suggested that T helper 17 (Th17) cells play a central role in the pathogenesis of ocular immune disease. The association between pathogenic Th17 cells and the development of uveitis has been confirmed in experimental and clinical studies. Several cytokines affect the initiation and stabilization of the differentiation of Th17 cells. Therefore, understanding the mechanism of related cytokines in the differentiation of Th17 cells is important for exploring the pathogenesis and the potential therapeutic targets of uveitis. This article briefly describes the structures, mechanisms, and targeted drugs of cytokines-including interleukin (IL)-6, transforming growth factor-β1 (TGF-β1), IL-1β, IL-23, IL-27, IL-35, IL-2, IL-4, IL-21, and interferon (IFN)-γ-which have an important influence on the differentiation of Th17 cells and discusses their potential as therapeutic targets for treating autoimmune uveitis.
Collapse
Affiliation(s)
- Kailei Guo
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Xiaomin Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| |
Collapse
|