1
|
Liu S, Yang X, Zhao H, Zhao X, Fan K, Liu G, Li X, Du C, Liu J, Ma J. Cathepsin C exacerbates EAE by promoting the expansion of Tfh cells and the formation of TLSs in the CNS. Brain Behav Immun 2025; 123:123-142. [PMID: 39243987 DOI: 10.1016/j.bbi.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/05/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) mediated by CD4+ T helper (Th) cells, and characterized by immune cell infiltration, demyelination and neurodegeneration, with no definitive cure available. Thus, it is pivotal and imperative to acquire more profound comprehension of the underlying mechanisms implicated in MS. Dysregulated immune responses are widely believed to play a primary role in the pathogenesis of MS. Recently, a plethora of studies have demonstrated the involvement of T follicular helper (Tfh) cells and tertiary lymphoid-like structures (TLSs) in the pathogenesis and progression of MS. Cathepsin C (CatC) is a cysteine exopeptidase which is crucial for the activation of immune-cell-associated serine proteinases in many inflammatory diseases in peripheral system, such as rheumatoid arthritis and septicemia. We have previously demonstrated that CatC is involved in neuroinflammation and exacerbates demyelination in both cuprizone-induced and experimental autoimmune encephalomyelitis (EAE) mouse models. However, the underlying immunopathological mechanism remains elusive. In the present study, we established a recombinant myelin oligodendrocyte glycoprotein 35-55 peptide-induced EAE model using conditional CatC overexpression mice to investigate the effects of CatC on the alteration of CD4+ Th subsets, including Th1, Th2, Th17, Tfh and T regulatory cells. Our findings demonstrated that CatC particularly enhanced the population of Tfh cell in the brain, resulting in the earlier onset and more severe chronic syndrome of EAE. Furthermore, CatC promoted the formation of TLSs in the brain, leading to persistent neuroinflammation and exacerbating the severity of EAE in the chronic phase. Conversely, treatment with AZD7986, a specific inhibitor of CatC, effectively attenuated the syndrome of EAE and its effects caused by CatC both in vivo and in vitro. These findings provide a novel insight into the critical role of CatC in innate and adaptive immunity in EAE, and specific inhibitor of CatC, AZD7986, may contribute to potential therapeutic strategies for MS.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xiaohan Yang
- Department of Morphology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Henan Zhao
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xinnan Zhao
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Kai Fan
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Gang Liu
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xia Li
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Cong Du
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Jianmei Ma
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China; National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China.
| |
Collapse
|
2
|
Ramzi M, Dehghani M, Hajimaghsoodi M, Golmoghaddam H, Arandi N. The impact of PD-1/PD-L1, CTLA-4, TIM-3 and LAG-3 immune checkpoint receptor expression in the development of acute graft versus host disease (aGVHD) and disease recurrence after allogeneic hematopoietic stem cell transplantation. Hum Immunol 2024; 86:111225. [PMID: 39740301 DOI: 10.1016/j.humimm.2024.111225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/26/2024] [Accepted: 12/18/2024] [Indexed: 01/02/2025]
Abstract
The immune checkpoint receptors play a crucial role in managing the transplantation outcome including development of acute graft versus host disease (aGVHD) and disease recurrence following allogeneic hematopoietic stem cell transplantation (allo-HSCT) is well established. This study aimed to investigate the expression of immune checkpoint receptors, including PD-1/PD-L1, CTLA-4, TIM-3, and LAG-3 in donors, as well as changes in their expression during the first 90 days (day 30 and day 90) post-HLA-matched allo-HSCT, concerning the development of aGVHD and disease relapse. Forty-one donor/recipient pairs were included in this study. The relative expression of immune checkpoint receptors was measured using the SYBR Green Real-Time PCR method. There was no significant relationship between the expression of PD-1/PD-L1, CTLA-4, TIM-3, and LAG-3 immune checkpoint receptors in donors and the occurrence of aGVHD and disease relapse. Additionally, alterations in the expression of these receptors during the initial 90 days post-transplantation did not correlate with aGVHD development. However, patients exhibiting elevated PD-L1 levels at day 90 had an increased risk of disease recurrence post-allo-HSCT (*P = 0.027). This study is the first to demonstrate that high PD-L1 expression in the peripheral blood at day 90 after allo-HSCT is associated with an increased rate of post-transplantation relapse.
Collapse
Affiliation(s)
- Mani Ramzi
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Stem Cell Transplantation, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dehghani
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Stem Cell Transplantation, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Hossein Golmoghaddam
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nargess Arandi
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Huang Z, Wang L, Li W, Liao N, Heng J, Qin Y, Li L, Bian Z, Cao W, Xia L, Zhang R. The role of lncRNA NEAT1 in acute graft-versus-host disease: Regulation of macrophage polarization and inflammatory cytokine secretion via JNK/NLRP3 pathway. Int Immunopharmacol 2024; 146:113857. [PMID: 39721453 DOI: 10.1016/j.intimp.2024.113857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 11/23/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Acute graft-versus-host disease (aGVHD) is a complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). The role of macrophages as proficient antigen-presenting cells in aGVHD is a prominent area of investigation in contemporary research. The association between long noncoding RNA nuclear enriched abundant transcript 1 (lncRNA NEAT1) and the macrophage function is of significant interest. However, the role of lncRNA NEAT1 in aGVHD needs to be further explored. METHODS Peripheral blood mononuclear cells (PBMCs) were collected from patients with or without aGVHD (non-aGVHD) after allo-HSCT. RAW264.7 cells and bone marrow-derived macrophages (BMDMs) were transduced with NEAT1 lentiviral vector or transfected with NEAT1 small interfering RNA to change the expression level of lncRNA NEAT1. Finally, an aGVHD mouse model was established to evaluate the role of JNK inhibitor or NLRP3 inhibitor in aGVHD. RESULTS Compared with non-aGVHD patients, lncRNA NEAT1 was significantly up-regulated in the PBMCs of aGVHD patients. ROC and AUC analysis confirmed that the expression of lncRNA NEAT1 was correlated with the occurrence of aGVHD. The overexpression of lncRNA NEAT1 in RAW264.7 could significantly promote the proliferation, migration, and differentiation into M1 macrophages. Knockdown of lncRNA NEAT1 could significantly decrease the proportion of M1 macrophages, regulate pro-inflammatory cytokines secretion, and affect the JNK/NLRP3 pathway in lipopolysaccharides (LPS)-induced BMDMs. Correspondingly, JNK and NLRP3 inhibitors reduced LPS-induced pro-inflammatory responses in macrophages. Furthermore, JNK and NLRP3 inhibitors regulated macrophage polarization and improved symptoms in aGVHD mice. CONCLUSIONS The aforementioned data suggest that lncRNA NEAT1 potentially plays a significant role in macrophage polarization and the secretion of inflammatory cytokines through its modulation of the JNK/NLRP3 pathway. Consequently, this study establishes a foundation for the development of novel therapeutic approaches targeting aGVHD.
Collapse
Affiliation(s)
- Zhenli Huang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ni Liao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingjing Heng
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Qin
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhilei Bian
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weijie Cao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Linghui Xia
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ran Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
4
|
Zhang A, Huang Z, Zhang R, Wei R, Jiang S, Chen H, Cao X, Shi W, Xia L, Hu Y. Humanized anti-CD25 monoclonal antibody replaces methotrexate as acute graft-versus-host disease prophylaxis in haploidentical allogeneic haematopoietic stem cell transplantation. Br J Haematol 2024. [PMID: 39710371 DOI: 10.1111/bjh.19958] [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: 09/19/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Acute graft-versus-host disease (aGVHD) significantly affects quality of life and outcomes in patients post-haploidentical haematopoietic stem cell transplantation (haplo-HSCT). Methotrexate (MTX) is commonly used to prevent aGVHD but can lead to complications like delayed haematological recovery and oral mucositis (OM). This study investigates the efficacy of anti-CD25 monoclonal antibody (mAb) as a potential MTX alternative. Participants were divided into two cohorts: a single-dose group (25 mg/day anti-CD25 mAb with MTX) and a double-dose group (50 mg/day anti-CD25 mAb without MTX). The primary end-point was the cumulative incidence (CI) of severe aGVHD by day 100. The double-dose cohort demonstrated a significantly lower CI of total aGVHD (23.53% vs. 42.11%, p = 0.009) and grade 3-4 aGVHD (7.35% vs. 18.42%, p = 0.047). After inverse probability of treatment weighting adjustment, the adjusted HR of double-dose compared with single-dose cohort for total aGVHD was 0.47 (95% CI 0.26-0.86; p = 0.015), 0.42(95% CI 0.15-1.22; p = 0.110) for grade III-IV aGVHD, 0.45 (95% CI 0.26-0.77; p = 0.004) for total cGVHD and 0.36 (95% CI 0.18-0.72; p = 0.004) for the moderate to severe cGVHD. Additionally, this double-dose regimen significantly reduced the incidence of oral mucositis and demonstrated lower rates of infections and haemorrhagic cystitis. These findings suggest that a double-dose anti-CD25 mAb regimen without MTX is a promising strategy for aGVHD prophylaxis in haplo-HSCT (ChiCTR2200060184).
Collapse
Affiliation(s)
- Ao Zhang
- Institute of Hematology, Union Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenli Huang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ran Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruowen Wei
- Institute of Hematology, Union Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Jiang
- Institute of Hematology, Union Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongru Chen
- Institute of Hematology, Union Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiena Cao
- Institute of Hematology, Union Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Shi
- Institute of Hematology, Union Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Linghui Xia
- Institute of Hematology, Union Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Luo J, Zhou Y, Wang M, Zhang J, Jiang E. Inflammasomes: potential therapeutic targets in hematopoietic stem cell transplantation. Cell Commun Signal 2024; 22:596. [PMID: 39695742 DOI: 10.1186/s12964-024-01974-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/30/2024] [Indexed: 12/20/2024] Open
Abstract
The realm of hematopoietic stem cell transplantation (HSCT) has witnessed remarkable advancements in elevating the cure and survival rates for patients with both malignant and non-malignant hematologic diseases. Nevertheless, a considerable number of patients continue to face challenges, including transplant-related complications, infection, graft failure, and mortality. Inflammasomes, the multi-protein complexes of the innate immune system, respond to various danger signals by releasing inflammatory cytokines and even mediating cell death. While moderate activation of inflammasomes is essential for immune defense and homeostasis maintenance, excessive activation precipitates inflammatory damage. The intricate interplay between HSCT and inflammasomes arises from their pivotal roles in immune responses and inflammation. This review examines the molecular architecture and composition of various types of inflammasomes, highlighting their activation and effector mechanisms within the context of the HSCT process and its associated complications. Additionally, we summarize the therapeutic implications of targeting inflammasomes and related factors in HSCT.
Collapse
Affiliation(s)
- Jieya Luo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yunxia Zhou
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Haihe Laboratory of Cell Ecosystem, Tianjin Medical University, Tianjin, 300051, China
| | - Mingyang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Junan Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
6
|
Shengchao M, Bo T, Huihui L, Chenchen Q, Beichen L, Zhenhua W, Ning M, Yongjin S. Long-term CXCR3 antagonist AMG487 mitigated acute graft-versus-host disease by inhibiting T cell activation in a murine model. Transpl Immunol 2024; 87:102128. [PMID: 39260677 DOI: 10.1016/j.trim.2024.102128] [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: 12/26/2023] [Revised: 09/04/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Lymphocyte migration plays a key role in the development of acute graft-versus-host disease (aGVHD). Blocking lymphocyte migration by targeting chemokine receptors, such as CXCR3, may be a promising strategy for preventing and treating aGVHD. Our previous studies have shown that short-term CXCR3 antagonist treatment combined with cyclosporine A alleviated aGVHD. However, the effect of long-term AMG487 treatment on aGVHD survival has not been thoroughly investigated. METHODS A murine aGVHD model was used to examine the expression of CXCR3 in donor T cells. The effects of short- and long-term AMG487 treatment on aGVHD survival were assessed. The infiltration of donor T cells into the liver and spleen tissues and the activation of donor T cells in splenic tissues were also examined. RESULTS CXCR3 was consistently highly expressed in donor T cells in a murine aGVHD model. Long-term AMG487 treatment, but not short-term, improved survival and aGVHD outcomes (p < 0.05). Furthermore, long-term AMG487 administration reduced the number of donor T cells in the liver but increased the number of donor T cells in the spleen (p < 0.05). Long-term AMG487 treatment also inhibited donor T cell activation in the spleen (p < 0.05). CONCLUSION This study demonstrates that long-term AMG487 treatment has a potential therapeutic effect on aGVHD and could be used as a novel therapy.
Collapse
Affiliation(s)
- Miao Shengchao
- Department of Hematology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.
| | - Tang Bo
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Liu Huihui
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Qin Chenchen
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Liu Beichen
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Wang Zhenhua
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Ma Ning
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Shi Yongjin
- Department of Hematology, Peking University First Hospital, Beijing, China
| |
Collapse
|
7
|
Öztürk G, Bayrakoğlu D, Haskoloğlu Ş, Baskın K, Deveci N, İnce E, İleri T, Çakmaklı H, Ertem M, İkincioğulları A, Doğu F. ST2 and Reg3α: Can they predict aGvHD, steroid refractoriness and transplant-related mortality in pediatric patients after HSCT? Hematol Transfus Cell Ther 2024; 46 Suppl 6:S129-S135. [PMID: 38658297 DOI: 10.1016/j.htct.2024.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 12/18/2023] [Accepted: 02/27/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND/AIM There are several complications of hematopoietic stem cell transplantation. Without any doubt, most important of these is aGvHD that increases transplant-related mortality. The aim of this study is to investigate whether ST-2 and Reg3α levels measured at an early stage in pediatric patients undergoing allogeneic hematopoietic stem cell transplantation can be individual biomarkers identifying future GvHD and predicting treatment response. MATERIALS AND METHODS From January 2019 to January 2021, 27 patients undergoing hematopoietic stem cell transplantation for primary immunodeficiency or hematopoietic diseases formed the study group. During their follow-up, the patients were classified into two groups as those developing and those not developing aGvHD. Nineteen healthy volunteers from a similar age group who needed their blood samples drawn for other reasons and who did not have any history of chronic disease, infection or medication use formed the control group. Blood samples of patients scheduled to have allogeneic HSCT were obtained before the administration of the preparative regimen, on Day +7 post-transplant and on the day of diagnosis if they developed aGvHD. Serum samples were stored at -20ºC until the day of processing. ST2 and Reg3α levels were measured using the ELISA method. RESULTS For patients who developed aGvHD (n = 13), ST2 levels obtained before the transplantation, on Day +7 post-transplant and on the day of aGvHD diagnosis (in patients developing GvHD) were significantly higher compared to the healthy Control Group (p-value <0.05). As regards to the samples obtained on the same days, ST2 levels did not differ significantly among patients who developed and those who did not develop GvHD (n = 14; p-value >0.05). ST2 levels of samples obtained on the days that acute skin and gastrointestinal tract GvHD developed did not differ significantly between these two groups (p-value >0.05). Reg3α levels of the pre-transplant samples, on Day +7 after the transplantation and on the day of aGvHD diagnosis did not show any difference between any of the groups (p-value >0.05). As only two patients died after transplantation, thus correlation of ST2 and Reg3α levels with transplant-related mortality could not be proven. CONCLUSION The results of this study suggest that ST2 and Reg3α levels are neither diagnostic nor prognostic or predictive biomarkers of aGvHD, steroid resistance or transplant-related mortality in pediatric patients. This study can be regarded as a pilot study because of the small patient population; more research involving a larger patient population is required.
Collapse
Affiliation(s)
- Gökcan Öztürk
- Ankara University School of Medicine, Department of Pediatric Immunology and Allergy, Ankara, Turkey.
| | - Deniz Bayrakoğlu
- Ankara University School of Medicine, Department of Pediatric Immunology and Allergy, Ankara, Turkey
| | - Şule Haskoloğlu
- Ankara University School of Medicine, Department of Pediatric Immunology and Allergy, Ankara, Turkey
| | - Kübra Baskın
- Ankara University School of Medicine, Department of Pediatric Immunology and Allergy, Ankara, Turkey
| | - Nazlı Deveci
- Ankara University School of Medicine, Department of Pediatric Immunology and Allergy, Ankara, Turkey
| | - Elif İnce
- Ankara University School of Medicine, Department of Pediatric Hematology, Ankara, Turkey
| | - Talia İleri
- Ankara University School of Medicine, Department of Pediatric Hematology, Ankara, Turkey
| | - Hasan Çakmaklı
- Ankara University School of Medicine, Department of Pediatric Hematology, Ankara, Turkey
| | - Mehmet Ertem
- Ankara University School of Medicine, Department of Pediatric Hematology, Ankara, Turkey
| | - Aydan İkincioğulları
- Ankara University School of Medicine, Department of Pediatric Hematology, Ankara, Turkey
| | - Figen Doğu
- Ankara University School of Medicine, Department of Pediatric Immunology and Allergy, Ankara, Turkey
| |
Collapse
|
8
|
Shi XZ, Zhang XY, Wang YY, Zhao YM, Wang J. Polysaccharides from Hericium erinaceus and its immunomodulatory effects on RAW 264.7 macrophages. Int J Biol Macromol 2024; 278:134947. [PMID: 39173803 DOI: 10.1016/j.ijbiomac.2024.134947] [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: 03/04/2024] [Revised: 08/10/2024] [Accepted: 08/20/2024] [Indexed: 08/24/2024]
Abstract
This study aimed to optimize the extraction of Hericium erinaceus polysaccharides (HEP) using ultrasound-assisted enzymatic extraction combined with Plackett-Burman design (PBD) and response surface methodology (RSM). The optimal extraction conditions were identified as: 33 min extraction time, 30:1 liquid to material ratio, 38 °C extraction temperature, 9 g/kg cellulase amount, pH 4, and 20 % ethanol concentration. Under these conditions, the extraction yield of HEP was 5.87 ± 0.16 %, consistent with the predicted results. Additionally, the potential immunomodulatory activity of HEP on RAW 264.7 macrophage was evaluated. The results revealed that HEP improved the immunostimulatory activity of RAW264.7 cells, evident from increased production of IL-6 and TNF-α. These findings suggest that HEP is capable of enhancing the immune activity of RAW 264.7 macrophage.
Collapse
Affiliation(s)
- Xiao-Zi Shi
- Department of Pharmacy, Hebei North University, Zhangjiakou, China
| | - Xin-Yan Zhang
- Department of Pharmacy, Hebei North University, Zhangjiakou, China; Tianjin Beichen Traditional Chinese Medicine Hospital
| | - Yin-Yue Wang
- Department of Pharmacy, Hebei North University, Zhangjiakou, China
| | - Yong-Ming Zhao
- Department of Pharmacy, Hebei North University, Zhangjiakou, China.
| | - Jin Wang
- Department of Pharmacy, Hebei North University, Zhangjiakou, China; Hebei Key Laboratory of Neuropharmacology, Zhangjiakou, China.
| |
Collapse
|
9
|
Sun H, Wu L, Zhao X, Huo Y, Dong P, Pang A, Zheng Y, Han Y, Ma S, Jiang E, Dong F, Cheng T, Hao S. Monocytes as an early risk factor for acute graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Front Immunol 2024; 15:1433091. [PMID: 39328417 PMCID: PMC11424452 DOI: 10.3389/fimmu.2024.1433091] [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: 05/15/2024] [Accepted: 08/13/2024] [Indexed: 09/28/2024] Open
Abstract
Acute graft-versus-host disease (aGVHD) is a major complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT) and contributes to high morbidity and mortality. However, our current understanding of the development and progression of aGVHD after allo-HSCT remains limited. To identify the potential biomarkers for the prevention and treatment of aGVHD during the early hematopoietic reconstruction after transplantation, we meticulously performed a comparative analysis of single-cell RNA sequencing data from post-transplant patients with or without aGVHD. Prior to the onset of aGVHD, monocytes in the peripheral blood of patients with aGVHD experienced a dramatic rise and activation on day 21 post-transplantation. This phenomenon is closely aligned with clinical cohort results obtained from blood routine examinations. Furthermore, in vitro co-culture experiments showed that peripheral blood monocytes extracted from patients with aGVHD approximately 21 days post-transplantation induced a significantly higher proliferation rate of allogeneic T cells compared to those from patients without aGVHD. Our study indicates that monocytes could be a crucial early clinical risk factor for the development of aGVHD, and this insight could potentially guide the timing of monitoring efforts, recommending assessments at the pivotal juncture of approximately day 21 post-transplantation, shedding fresh light on the significance of early hematopoietic regeneration in relation to the onset of aGVHD.
Collapse
Affiliation(s)
- Huimin Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Linjie Wu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xueying Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yingying Huo
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Peiyuan Dong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Aiming Pang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yawei Zheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yiwen Han
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Shihui Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Fang Dong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Sha Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
10
|
Chen F, Sheng J, Li X, Gao Z, Hu L, Chen M, Fei J, Song Z. Tumor-associated macrophages: orchestrators of cholangiocarcinoma progression. Front Immunol 2024; 15:1451474. [PMID: 39290697 PMCID: PMC11405194 DOI: 10.3389/fimmu.2024.1451474] [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: 06/19/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a rare but highly invasive cancer, with its incidence rising in recent years. Currently, surgery remains the most definitive therapeutic option for CCA. However, similar to other malignancies, most CCA patients are not eligible for surgical intervention at the time of diagnosis. The chemotherapeutic regimen of gemcitabine combined with cisplatin is the standard treatment for advanced CCA, but its effectiveness is often hampered by therapeutic resistance. Recent research highlights the remarkable plasticity of tumor-associated macrophages (TAMs) within the tumor microenvironment (TME). TAMs play a crucial dual role in either promoting or suppressing tumor development, depending on the factors that polarize them toward pro-tumorigenic or anti-tumorigenic phenotypes, as well as their interactions with cancer cells and other stromal components. In this review, we critically examine recent studies on TAMs in CCA, detailing the expression patterns and prognostic significance of different TAM subtypes in CCA, the mechanisms by which TAMs influence CCA progression and immune evasion, and the potential for reprogramming TAMs to enhance anticancer therapies. This review aims to provide a framework for deeper future research.
Collapse
Affiliation(s)
- Fei Chen
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jian Sheng
- Department of Research and Teaching, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xiaoping Li
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Zhaofeng Gao
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Lingyu Hu
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Minjie Chen
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jianguo Fei
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Zhengwei Song
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
11
|
Su L, Wei ZF, Pi CC, Qin TX, Song F, Zhang YW, Gao SJ. Icariin protects against acute graft-versus-host disease while preserving graft-versus-leukemia activity after allogeneic hematopoietic stem cell transplantation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155901. [PMID: 39067193 DOI: 10.1016/j.phymed.2024.155901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/27/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Acute graft-versus-host disease (aGVHD), which is mainly mediated by allogeneic T cells, is a decisive factor in the success of allogeneic hematopoietic stem cell transplantation (allo-HCT). Prophylaxis for aGVHD in clinical patients is unsatisfactory, and there is still a huge unmet need for novel approaches. Icariin (ICA) shows potent anti-inflammatory activity and suppresses T cell-mediated immune responses. Thus, ICA is a potential drug for the prevention of aGVHD. However, there is no data assessing the impact of ICA on aGVHD after allo-HCT. PURPOSE This study aimed to investigate the protective effect of ICA against aGVHD and its mechanisms. Moreover, the impact of ICA on the graft-versus-leukemia (GVL) effect and engraftment of donor hematopoietic and immune cells were assessed. METHODS Different murine models of allo-HCT were developed to study the influence of the ICA on GVHD and GVL effect. Flow cytometry was used to analyze the growth of leukemia cells, alterations in different immune cells, and apoptosis. Cell proliferation was determined using a CCK-8 assay. RNA sequencing and quantitative proteomic analysis were performed to elucidate the underlying mechanisms, which were further verified by polymerase chain reaction or functional experiments. RESULTS Different concentrations of ICA exhibited opposite effects: low-concentration ICA promoted, while high concentrations suppressed the proliferation and function of T cells. A high dose of ICA administration during days +3 to +5 post-allo-HCT can alleviate murine aGVHD but does not affect the course of chronic GVHD (cGVHD), the GVL effect against both acute myeloid and lymphoblastic leukemia, or the recovery of donor hematological and immune cells. ICA extensively represses the expansion, function, and infiltration of donor alloreactive T cells, while preserving regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSC). Quantitative proteomic analysis showed that downregulation of integrin-linked kinase (ILK) and lymphocyte cytosolic protein 2 (LCP2) expression was possibly associated with ICA-mediated aGVHD protective effects. Furthermore, an inhibitor of ILK, which can alleviate murine aGVHD administered early after allo-HCT. CONCLUSION These findings suggest that the bioactivities of ICA are associated with its concentration and that ICA can effectively mitigate aGVHD without losing GVL activity or engraftment of donor hematopoietic and immune cells. Thus, ICA may be a promising drug for preventing aGVHD in clinical settings.
Collapse
Affiliation(s)
- Long Su
- Department of Hematology, The First Hospital of Jilin University, Changchun 130021, China; Key Laboratory of Hematology Precision Medicine of Jilin Province, The First Hospital of Jilin University, Changchun 130021, China
| | - Zhi-Feng Wei
- Department of Hematology, The First Hospital of Jilin University, Changchun 130021, China; Key Laboratory of Hematology Precision Medicine of Jilin Province, The First Hospital of Jilin University, Changchun 130021, China
| | - Chen-Chen Pi
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun 130061, China
| | - Tian-Xue Qin
- Department of Hematology, The First Hospital of Jilin University, Changchun 130021, China; Key Laboratory of Hematology Precision Medicine of Jilin Province, The First Hospital of Jilin University, Changchun 130021, China
| | - Fei Song
- Department of Hematology, The First Hospital of Jilin University, Changchun 130021, China; Key Laboratory of Hematology Precision Medicine of Jilin Province, The First Hospital of Jilin University, Changchun 130021, China
| | - Yun-Wei Zhang
- Department of Hematology, The First Hospital of Jilin University, Changchun 130021, China; Key Laboratory of Hematology Precision Medicine of Jilin Province, The First Hospital of Jilin University, Changchun 130021, China
| | - Su-Jun Gao
- Department of Hematology, The First Hospital of Jilin University, Changchun 130021, China; Key Laboratory of Hematology Precision Medicine of Jilin Province, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
12
|
Singh RB, Cho W, Liu C, Naderi A, Surico PL, Kahale F, Dohlman TH, Chauhan SK, Dana R. Immunopathological mechanisms and clinical manifestations of ocular graft-versus-host disease following hematopoietic stem cell transplantation. Bone Marrow Transplant 2024; 59:1049-1056. [PMID: 38822141 DOI: 10.1038/s41409-024-02321-3] [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: 03/30/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
Graft-versus-host disease is among the most common clinical complications following allogeneic hematopoietic stem cell transplantation. It causes inflammation-mediated destruction and dysfunction of various organ systems including ocular tissues in 60-90% of the patients and is termed ocular GVHD (oGVHD). In oGVHD, donor-derived T-cells recognize host antigens as foreign, resulting in immune dysregulation, inflammation and fibrosis of lacrimal glands, meibomian glands, cornea, and conjunctiva. The clinical presentation in oGVHD patients range from mild dry eye symptoms to catastrophic inflammation mediated pathological changes which can cause corneal perforation and blindness. In this review article, we provide detailed insights into the impact of mucosal barrier disruption, the afferent and efferent phases of immunological response involving activation of antigen presenting cells and T cells, respectively. We evaluate the evidence outlining the effector phase of the disease leading to cellular destruction and eventually fibrosis in patients with oGVHD. Finally, we discuss the well-established criteria for the diagnosis of oGVHD.
Collapse
Affiliation(s)
- Rohan Bir Singh
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Wonkyung Cho
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Catherine Liu
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Amirreza Naderi
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Pier Luigi Surico
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Francesca Kahale
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Thomas H Dohlman
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Sunil K Chauhan
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Reza Dana
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Liu J, Zhou Q, Meng K, Yang X, Ma B, Su C, Duan X. Aspirin Inhibits Colorectal Cancer via the TIGIT-BCL2-BAX pathway in T Cells. Int J Med Sci 2024; 21:1990-1999. [PMID: 39113892 PMCID: PMC11302567 DOI: 10.7150/ijms.98343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
The T cell immunoglobulin and ITAM domain (TIGIT) is a recently discovered synergistic co-suppressor molecule that plays an important role in immune response and tumor immune escape in the context of cancer. Importantly, CD155 acts as a receptor for TIGIT, and CD155 signaling to immune cells is mediated through interactions with the co-stimulatory immune receptor CD226 (DNAM-1) and the inhibitory checkpoint receptors TIGIT and CD96. Aspirin (ASA) has been shown to reduce the growth and survival of colorectal cancer (CRC) cells, but the immunological mechanisms involved have not been sufficiently elucidated. In the present study the effects of aspirin on CRC in mice and on Jurkat cells were investigated. Aspirin may suppress the expression of TIGIT on T cells and Regulatory T cells (Tregs) and inhibit T cell viability, and therefore induce tumor cell apoptosis. TIGIT is expressed at higher levels on infiltrating lymphocytes within CRC tumor tissue than adjacent. Further, aspirin could inhibit Jurkat cell proliferation and induce apoptosis via downregulation of TIGIT expression and the anti-apoptosis B cell lymphoma 2 (BCL2) protein and upregulation of BCL2-associated X protein (BAX) expression. The present study suggests that aspirin can inhibit specific aspects of T cell function by reducing interleukin-10 and transforming growth factor-β1 secretion via the TIGIT-BCL2-BAX signaling pathway, resulting in improved effector T cell function that inhibits tumor progression.
Collapse
Affiliation(s)
- Jiayu Liu
- School of Inspection, Ningxia Medical University, Yinchuan 750004, China
- The First School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Qiunan Zhou
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750004, China
| | - Kai Meng
- Department of Pathogen Biology and Immunology, School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
- Traditional Chinese Medicine Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Xiaojuan Yang
- School of Inspection, Ningxia Medical University, Yinchuan 750004, China
| | - Bin Ma
- Department of Oncology Surgery, The First People's Hospital of Yinchuan, Yinchuan 750004, China
| | - Chunxia Su
- Department of Pathogen Biology and Immunology, School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| | - Xiangguo Duan
- School of Inspection, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
14
|
Huang P, Xu J, Jiang S, Zhang Y, Wang X, Xiong C, Xia C. The impact of ICOSL/ICOS pathway-regulated long non-coding RNAs on liver fibrosis in mice infected with Schistosoma japonicum. Parasit Vectors 2024; 17:317. [PMID: 39044218 PMCID: PMC11267842 DOI: 10.1186/s13071-024-06399-y] [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: 05/15/2024] [Accepted: 07/08/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND The primary pathogenic mechanism of schistosomiasis-associated liver fibrosis involves the deposition of schistosome eggs, leading to the formation of liver egg granulomas and subsequent liver fibrosis. Hepatic stellate cells are abnormally activated, resulting in excessive collagen deposition and fibrosis development. While specific long non-coding RNAs (lncRNAs) have been associated with fibrotic processes, their roles in schistosomiasis-associated liver fibrosis remain unclear. METHODS Our previous research indicated that downregulating the ICOSL/ICOS could partially alleviate liver fibrosis. In this study, we established a schistosomiasis infection model in C57BL/6 and ICOSL knockout (KO) mice, and the liver pathology changes were observed at various weeks postinfection (wpi) using hematoxylin and eosin and Masson's trichrome staining. Within the first 4 wpi, no significant liver abnormalities were observed. However, mice exhibited evident egg granulomas and fibrosis in their livers at 7 wpi. Notably, ICOSL-KO mice had significantly smaller pathological variations compared with simultaneously infected C57BL/6 mice. To investigate the impact of lncRNAs on schistosomiasis-associated liver fibrosis, quantitative real-time polymerase chain reaction (RT-qPCR) was used to monitor the dynamic changes of lncRNAs in hepatic stellate cells of infected mice. RESULTS The results demonstrated that lncRNA-H19, -MALAT1, -PVT1, -P21 and -GAS5 all participated in liver fibrosis formation after schistosome infection. In addition, ICOSL-KO mice exhibited significantly inhibited expression of lncRNA-H19, -MALAT1 and -PVT1 after 7 wpi. In contrast, they showed enhanced expression of lncRNA-P21 and -GAS5 compared with C57BL/6 mice, influencing liver fibrosis development. Furthermore, small interfering RNA transfection (siRNA) in JS-1 cells in vitro confirmed that lncRNA-H19, -MALAT1, and -PVT1 promoted liver fibrosis, whereas lncRNA-P21 and -GAS5 had the opposite effect on key fibrotic molecules, including α- smooth muscle actin and collagen I expression. CONCLUSIONS This study uncovers that ICOSL/ICOS may play a role in activating hepatic stellate cells and promoting liver fibrosis in mice infected with Schistosoma japonicum by dynamically regulating the expression of specific lncRNAs. These findings offer potential therapeutic targets for schistosomiasis-associated liver fibrosis.
Collapse
Affiliation(s)
- Ping Huang
- Department of Parasitology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Jing Xu
- Department of Parasitology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Suqin Jiang
- Department of Parasitology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Yanan Zhang
- Department of Parasitology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Xinyi Wang
- Department of Parasitology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Chunrong Xiong
- Jiangsu Institute of Parasitic Diseases, Wuxi City, Jiangsu Province, China
| | - Chaoming Xia
- Department of Parasitology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China.
| |
Collapse
|
15
|
Huang K, Yang M, Zhou Y, Cao Y, Pang G, Zhao J, Liu Y, Luo J. Application of CD25 and CTLA4 gene transcription levels in early prediction of acute graft-versus-host disease. Front Immunol 2024; 15:1410439. [PMID: 39072333 PMCID: PMC11272456 DOI: 10.3389/fimmu.2024.1410439] [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: 04/01/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Our study investigated the potential of peripheral blood T cell CD25, CD28, and CTLA-4 gene transcription levels as predictive biomarkers for acute graft-versus-host disease (aGVHD) following allogeneic hematopoietic stem cell transplantation (allo-HSCT). Methods Real-time reverse transcription fluorescent quantitative PCR (RT-qPCR) analysis was conducted on day +7, +14, and +21 post-transplantation in patients undergoing allo-HSCT. Results Elevated levels of CD25 and CTLA-4 mRNA were found to be associated with the occurrence of aGVHD, as well as severe and gastrointestinal aGVHD. Receiver operating characteristic (ROC) curve analysis was utilized to assess the predictive value of each biomarker. Combined analysis of CD25 and CTLA-4 mRNA levels demonstrated promising predictive potential for aGVHD. Conclusion Our results confirmed that the transcription levels of CD25 and CTLA-4 genes could be used as early predictive biomarkers for aGVHD post-allo-HSCT.
Collapse
Affiliation(s)
- Ken Huang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Pediatrics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Mengxin Yang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yuhang Zhou
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Institute of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yaxuan Cao
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guanxiu Pang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jie Zhao
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yang Liu
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jianming Luo
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
16
|
Konuma T, Hamatani-Asakura M, Monna-Oiwa M, Kato S, Isobe M, Yokoyama K, Takahashi S, Nannya Y. Effect of IL-2 polymorphism rs2069762 on single-unit cord blood transplant outcomes. Cytokine 2024; 179:156636. [PMID: 38718489 DOI: 10.1016/j.cyto.2024.156636] [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: 02/12/2024] [Revised: 04/23/2024] [Accepted: 05/02/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Interleukin-2 (IL-2) is one of the most important cytokines that regulate the activation and proliferation of T cells and natural killer cells. The production of IL-2 may be affected by polymorphisms in the promoter region of the IL-2 gene (rs2069762). In allogeneic hematopoietic cell transplantation (HCT) from adult donors, rs2069762 has been associated with the incidence of acute and chronic graft-versus-host disease (GVHD). However, the impacts of IL-2 polymorphism on cord blood transplantation (CBT) outcomes remain unclear. OBJECTIVE The objective of this study was to assess the impact of IL-2 polymorphism rs2069762 on transplant outcomes, such as hematopoietic recovery, GVHD, overall survival, relapse, and non-relapse mortality (NRM) after CBT. STUDY DESIGN We conducted a retrospective analysis of data from adult patients who underwent single-unit CBT at our institution from November 2005 to March 2023 for whom DNA samples from recipients and donors were available. IL-2 genotyping was performed using real-time polymerase chain reaction with the TaqMan® SNP genotyping assay for rs2069762. RESULTS A total of 143 recipient and donor pairs were included in this study. The proportion of recipient IL-2 polymorphism rs2069762 was 48 % (n = 69) for AA, 42 % (n = 60) for CA, and 10 % (n = 14) for CC. The proportion of donor IL-2 polymorphism rs2069762 was 43 % (n = 61) for AA, 48 % (n = 69) for CA, and 9 % (n = 13) for CC. In the multivariate analysis, the use of an rs2069762 CA + CC donor was associated with lower neutrophil recovery compared to an rs2069762 AA donor (hazard ratio [HR], 0.66; 95 % confidence interval [CI], 0.50-0.88; P = 0.004). Furthermore, recipients of rs2069762 CA + CC were associated with higher NRM compared to recipients of rs2069762 AA (HR, 2.32; 95 % CI, 1.01-5.34; P = 0.047). Serum IL-2 levels at 8 weeks were significantly higher in rs2069762 CA + CC recipients compared to those with rs2069762 AA recipients (P = 0.014). CONCLUSION Our data showed that donor IL-2 polymorphism affects neutrophil recovery and recipient IL-2 polymorphism affects NRM in adults undergoing single-unit CBT. The polymorphism of IL-2 rs2069762 in recipients and donors might be associated with the clinical outcomes of single-unit CBT.
Collapse
Affiliation(s)
- Takaaki Konuma
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Megumi Hamatani-Asakura
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Maki Monna-Oiwa
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seiko Kato
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masamichi Isobe
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kazuaki Yokoyama
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoshi Takahashi
- Division of Clinical Precision Research Platform, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yasuhito Nannya
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
Jiang Z, Cai G, Liu H, Liu L, Huang R, Nie X, Gui R, Li J, Ma J, Cao K, Luo Y. A combination of a TLR7/8 agonist and an epigenetic inhibitor suppresses triple-negative breast cancer through triggering anti-tumor immune. J Nanobiotechnology 2024; 22:296. [PMID: 38811964 PMCID: PMC11134718 DOI: 10.1186/s12951-024-02525-1] [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: 01/15/2024] [Accepted: 05/02/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Combination therapy involving immune checkpoint blockade (ICB) and other drugs is a potential strategy for converting immune-cold tumors into immune-hot tumors to benefit from immunotherapy. To achieve drug synergy, we developed a homologous cancer cell membrane vesicle (CM)-coated metal-organic framework (MOF) nanodelivery platform for the codelivery of a TLR7/8 agonist with an epigenetic inhibitor. METHODS A novel biomimetic codelivery system (MCM@UN) was constructed by MOF nanoparticles UiO-66 loading with a bromodomain-containing protein 4 (BRD4) inhibitor and then coated with the membrane vesicles of homologous cancer cells that embedding the 18 C lipid tail of 3M-052 (M). The antitumor immune ability and tumor suppressive effect of MCM@UN were evaluated in a mouse model of triple-negative breast cancer (TNBC) and in vitro. The tumor immune microenvironment was analyzed by multicolor immunofluorescence staining. RESULTS In vitro and in vivo data showed that MCM@UN specifically targeted to TNBC cells and was superior to the free drug in terms of tumor growth inhibition and antitumor immune activity. In terms of mechanism, MCM@UN blocked BRD4 and PD-L1 to prompt dying tumor cells to disintegrate and expose tumor antigens. The disintegrated tumor cells released damage-associated molecular patterns (DAMPs), recruited dendritic cells (DCs) to efficiently activate CD8+ T cells to mediate effective and long-lasting antitumor immunity. In addition, TLR7/8 agonist on MCM@UN enhanced lymphocytes infiltration and immunogenic cell death and decreased regulatory T-cells (Tregs). On clinical specimens, we found that mature DCs infiltrating tumor tissues of TNBC patients were negatively correlated with the expression of BRD4, which was consistent with the result in animal model. CONCLUSION MCM@UN specifically targeted to TNBC cells and remodeled tumor immune microenvironment to inhibit malignant behaviors of TNBC.
Collapse
Affiliation(s)
- Zhenzhen Jiang
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Guangqing Cai
- Department of Orthopedics, Changsha Hospital of Traditional Chinese Medicine (Changsha Eighth Hospital), Changsha, Hunan, 410013, P. R. China
| | - Haiting Liu
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Leping Liu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Rong Huang
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Xinmin Nie
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Rong Gui
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Jian Li
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Jinqi Ma
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Ke Cao
- Department of Oncology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China.
| | - Yanwei Luo
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China.
| |
Collapse
|
18
|
Zhang P, Fleming P, Andoniou CE, Waltner OG, Bhise SS, Martins JP, McEnroe BA, Voigt V, Daly S, Kuns RD, Ekwe AP, Henden AS, Saldan A, Olver S, Varelias A, Smith C, Schmidt CR, Ensbey KS, Legg SR, Sekiguchi T, Minnie SA, Gradwell M, Wagenaar I, Clouston AD, Koyama M, Furlan SN, Kennedy GA, Ward ES, Degli-Esposti MA, Hill GR, Tey SK. IL-6-mediated endothelial injury impairs antiviral humoral immunity after bone marrow transplantation. J Clin Invest 2024; 134:e174184. [PMID: 38557487 PMCID: PMC10977988 DOI: 10.1172/jci174184] [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: 07/24/2023] [Accepted: 02/09/2024] [Indexed: 04/04/2024] Open
Abstract
Endothelial function and integrity are compromised after allogeneic bone marrow transplantation (BMT), but how this affects immune responses broadly remains unknown. Using a preclinical model of CMV reactivation after BMT, we found compromised antiviral humoral responses induced by IL-6 signaling. IL-6 signaling in T cells maintained Th1 cells, resulting in sustained IFN-γ secretion, which promoted endothelial cell (EC) injury, loss of the neonatal Fc receptor (FcRn) responsible for IgG recycling, and rapid IgG loss. T cell-specific deletion of IL-6R led to persistence of recipient-derived, CMV-specific IgG and inhibited CMV reactivation. Deletion of IFN-γ in donor T cells also eliminated EC injury and FcRn loss. In a phase III clinical trial, blockade of IL-6R with tocilizumab promoted CMV-specific IgG persistence and significantly attenuated early HCMV reactivation. In sum, IL-6 invoked IFN-γ-dependent EC injury and consequent IgG loss, leading to CMV reactivation. Hence, cytokine inhibition represents a logical strategy to prevent endothelial injury, thereby preserving humoral immunity after immunotherapy.
Collapse
Affiliation(s)
- Ping Zhang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Peter Fleming
- Infection and Immunity Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Christopher E. Andoniou
- Infection and Immunity Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Olivia G. Waltner
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Shruti S. Bhise
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jose Paulo Martins
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | | | - Valentina Voigt
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Sheridan Daly
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Rachel D. Kuns
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Adaeze P. Ekwe
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Andrea S. Henden
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- University of Queensland, St Lucia, Queensland, Australia
- Royal Brisbane and Women’s Hospital, Herston, Queensland, Australia
| | - Alda Saldan
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- University of Queensland, St Lucia, Queensland, Australia
| | - Stuart Olver
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- University of Queensland, St Lucia, Queensland, Australia
| | - Corey Smith
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Christine R. Schmidt
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Kathleen S. Ensbey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Samuel R.W. Legg
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Tomoko Sekiguchi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Simone A. Minnie
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Mark Gradwell
- Cancer Sciences Unit, Centre for Cancer Immunology, University of Southampton, Southampton, United Kingdom
| | - Irma Wagenaar
- Cancer Sciences Unit, Centre for Cancer Immunology, University of Southampton, Southampton, United Kingdom
| | | | - Motoko Koyama
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Scott N. Furlan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Pediatrics and
| | - Glen A. Kennedy
- University of Queensland, St Lucia, Queensland, Australia
- Royal Brisbane and Women’s Hospital, Herston, Queensland, Australia
| | - E Sally Ward
- Cancer Sciences Unit, Centre for Cancer Immunology, University of Southampton, Southampton, United Kingdom
| | - Mariapia A. Degli-Esposti
- Infection and Immunity Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Geoffrey R. Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Division of Medical Oncology, University of Washington, Seattle, Washington, USA
| | - Siok-Keen Tey
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- University of Queensland, St Lucia, Queensland, Australia
- Royal Brisbane and Women’s Hospital, Herston, Queensland, Australia
| |
Collapse
|
19
|
López-Andrade B, Cunill V, Andreu V, Bento L, Segura-Guerrero M, Moñino A, Iglesias J, Julià MR, Durán MA, Ballester MC, Muncunill J, Sampol A. Plasma from patients undergoing allogeneic hematopoietic stem cell transplantation promotes NETOSIS in vitro and correlates with inflammatory parameters and clinical severity. Front Immunol 2024; 15:1353106. [PMID: 38550584 PMCID: PMC10972998 DOI: 10.3389/fimmu.2024.1353106] [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: 12/09/2023] [Accepted: 02/28/2024] [Indexed: 04/02/2024] Open
Abstract
Introduction NETosis, the mechanism by which neutrophils release extracellular traps (NETs), is closely related to inflammation. During the allogeneic hematopoietic stem cell transplantation (allo-HSCT), different stimuli can induce NETs formation. Inflammation and endothelial injury have been associated with acute graft-versus-host disease (aGVHD) and complications after allo-HSCT. We focus on the study of NETosis and its relation with cytokines, hematological and biochemical parameters and clinical outcomes before, during and after allo-HSCT. Methods We evaluate the capacity of plasma samples from allo-HSCT patients to induce NETosis, in a cell culture model. Plasma samples from patients undergoing allo-HSCT had a stronger higher NETs induction capacity (NETsIC) than plasma from healthy donors throughout the transplantation process. An optimal cut-off value by ROC analysis was established to discriminate between patients whose plasma triggered NETosis (NETs+IC group) and those who did not (NETs-IC group). Results Prior to conditioning treatment, the capacity of plasma samples to trigger NETosis was significantly correlated with the Endothelial Activation and Stress Index (EASIX) score. At day 5 after transplant, patients with a positive NETsIC had higher interleukin (IL)-6 and C-reactive protein (CRP) levels and also a higher Modified EASIX score (M-EASIX) than patients with a negative NETsIC. EASIX and M-EASIX scores seek to determine inflammation and endothelium damage, therefore it could indicate a heightened immune response and inflammation in the group of patients with a positive NETsIC. Cytokine levels, specifically IL-8 and IL-6, significantly increased after allo-HSCT with peak levels reached on day 10 after graft infusion. Only, IL-10 and IL-6 levels were significantly higher in patients with a positive NETsIC. In our small cohort, higher IL-6 and IL-8 levels were related to early severe complications (before day 15 after transplant). Discussion Although early complications were not related to NETosis by itself, NETosis could predict overall non-specific but clinically significant complications during the full patient admission. In summary, NETosis can be directly induced by plasma from allo-HSCT patients and NETsIC was associated with clinical indicators of disease severity, cytokines levels and inflammatory markers.
Collapse
Affiliation(s)
- Bernardo López-Andrade
- Department of Hematology, Hospital Universitari Son Espases, Palma, Spain
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Vanesa Cunill
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Valero Andreu
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Leyre Bento
- Department of Hematology, Hospital Universitari Son Espases, Palma, Spain
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Marina Segura-Guerrero
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Andrea Moñino
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Julio Iglesias
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Maria Rosa Julià
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Maria Antonia Durán
- Department of Hematology, Hospital Universitari Son Espases, Palma, Spain
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Maria Carmen Ballester
- Department of Hematology, Hospital Universitari Son Espases, Palma, Spain
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Josep Muncunill
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Antonia Sampol
- Department of Hematology, Hospital Universitari Son Espases, Palma, Spain
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| |
Collapse
|
20
|
Huang Z, Zhang R, Teng Y, Guo J, Zhang H, Wang L, Tang LV, Shi W, Wu Q, Xia L. Nuclear Matrix-associated Protein SMAR1 Attenuated Acute Graft-versus-host Disease by Targeting JAK-STAT Signaling in CD4 + T Cells. Transplantation 2024; 108:e23-e35. [PMID: 37817309 DOI: 10.1097/tp.0000000000004818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2023]
Abstract
BACKGROUND Acute graft-versus-host disease (aGVHD) mediated by alloreactive T cells remains a serious and life-threatening complication of allogeneic hematopoietic cell transplantation (allo-HCT). The contribution of the different CD4 + T helper cell subtypes to the pathogenesis and regulation of aGVHD is a central point in current research. The specialized effector subsets of T cells that differentiate from naive T cells into mature cells are closely related to scaffold/matrix-associated region-1-binding protein (SMAR1). However, the role of SMAR1 in aGVHD is unclear. METHODS Peripheral blood was collected from the patients with or without aGVHD after allo-HCT. The differences in CD4 + T cells transduced with the SMAR1 lentivirus vector and empty vector were analyzed. A humanized aGVHD mouse model was constructed to evaluate the function of SMAR1 in aGVHD. RESULTS The expression of SMAR1 was significantly reduced in the CD4 + T cells from aGVHD patients and related to the occurrence of aGVHD. SMAR1 overexpression in human CD4 + T cells regulated CD4 + T-cell subsets differentiation and inflammatory cytokines secretion and inhibited the Janus kinase/signal transducer and activator of transcription pathway. Moreover, SMAR1 changed chromatin accessibility landscapes and affected the binding motifs of key transcription factors regulating T cells. Additionally, upregulation of SMAR1 expression in CD4 + T cells improved the survival and pathology in a humanized aGVHD mouse model. CONCLUSIONS Our results showed that upregulation of SMAR1 regulated the CD4 + T-cell subpopulation and cytokines secretion and improved survival in a humanized aGVHD mouse model by alleviating inflammation. This study provides a promising therapeutic target for aGVHD.
Collapse
Affiliation(s)
- Zhenli Huang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ran Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yao Teng
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Guo
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyong Zhang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liang V Tang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Shi
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuling Wu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linghui Xia
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Strobl J, Gail LM, Krecu L, Madad S, Kleissl L, Unterluggauer L, Redl A, Brazdilova K, Saluzzo S, Wohlfarth P, Knaus HA, Mitterbauer M, Rabitsch W, Haniffa M, Stary G. Diverse macrophage populations contribute to distinct manifestations of human cutaneous graft-versus-host disease. Br J Dermatol 2024; 190:402-414. [PMID: 38010706 PMCID: PMC10873647 DOI: 10.1093/bjd/ljad402] [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: 07/07/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Graft-versus-host disease (GvHD) is a major life-threatening complication of allogeneic haematopoietic stem cell transplantation (HSCT), limiting the broad application of HSCT for haematological malignancies. Cutaneous GvHD is described as a post-transplant inflammatory reaction by skin-infiltrating donor T cells and remaining recipient tissue-resident memory T cells. Despite the major influence of lymphocytes on GvHD pathogenesis, the complex role of mononuclear phagocytes (MNPs) in tissues affected by GvHD is increasingly appreciated. OBJECTIVES To characterize the identity, origin and functions of MNPs in patients with acute cutaneous GvHD. METHODS Using single-cell RNA sequencing and multiplex tissue immunofluorescence, we identified an increased abundance of MNPs in skin and blood from 36 patients with acute cutaneous GvHD. In cases of sex-mismatched transplantation, we used expression of X-linked genes to detect rapid tissue adaptation of newly recruited donor MNPs resulting in similar transcriptional states of host- and donor-derived macrophages within GvHD skin lesions. RESULTS We showed that cutaneous GvHD lesions harbour expanded CD163+ tissue-resident macrophage populations with anti-inflammatory and tissue-remodelling properties including interleukin-10 cytokine production. Cell-cell interaction analyses revealed putative signalling to strengthen regulatory T-cell responses. Notably, macrophage polarization in chronic cutaneous GvHD types was proinflammatory and drastically differed from acute GvHD, supporting the notion of distinct cellular players in different clinical GvHD subtypes. CONCLUSIONS Overall, our data reveal a surprisingly dynamic role of MNPs after HSCT. Specific and time-resolved targeting to repolarize this cell subset may present a promising therapeutic strategy in combatting GvHD skin inflammation.
Collapse
Affiliation(s)
- Johanna Strobl
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Laura M Gail
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Laura Krecu
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Shaista Madad
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- University of Cambridge, Cambridge, UK
| | - Lisa Kleissl
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Luisa Unterluggauer
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Anna Redl
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Kveta Brazdilova
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Simona Saluzzo
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Philipp Wohlfarth
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, 1090 Vienna, Austria
| | - Hanna A Knaus
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, 1090 Vienna, Austria
| | - Margit Mitterbauer
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, 1090 Vienna, Austria
| | - Werner Rabitsch
- Department of Internal Medicine I, Bone Marrow Transplantation Unit, Medical University of Vienna, 1090 Vienna, Austria
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| |
Collapse
|
22
|
Takano A, Oya K, Ishii Y, Maruyama Y, Nomura T. Amicrobial pustulosis of the folds concurrent with graft-versus-host disease. J Dermatol 2024; 51:e35-e36. [PMID: 37735973 DOI: 10.1111/1346-8138.16976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/14/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023]
Affiliation(s)
- Aya Takano
- Department of Dermatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kazumasa Oya
- Department of Dermatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yoshiyuki Ishii
- Department of Dermatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yumiko Maruyama
- Department of Hematology, University of Tsukuba Hospital, Ibaraki, Japan
| | - Toshifumi Nomura
- Department of Dermatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
23
|
Xie L, Ding Y, Zhang X. Melatonin and ovarian tissue transplantation: Current frontiers in research. J Gynecol Obstet Hum Reprod 2024; 53:102726. [PMID: 38219858 DOI: 10.1016/j.jogoh.2024.102726] [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: 04/13/2023] [Revised: 12/29/2023] [Accepted: 01/11/2024] [Indexed: 01/16/2024]
Abstract
The progress achieved in anticancer therapy in recent years has been paralleled by an increase in the survival of women with cancer globally. Nonetheless, the gonadotoxic impact of anticancer drugs has led to ovarian failure in treated women. While there are documented cases of successful ovarian tissue transplants resulting in restored fertility and childbirth, challenges persist, including suboptimal functional recovery and limited graft lifespan. Melatonin, an inert hormone primarily secreted by the mammalian pineal gland, exhibits diverse physiological functions, including antioxidative, anti-inflammatory, anti-apoptotic, and angiogenesis-regulating properties. Consequently, researchers have explored melatonin as a modulator to enhance graft function recovery in ovarian transplantation experiments, yielding promising outcomes. This review examines the relevant literature, consolidating findings that underscore the positive effects of melatonin in safeguarding the morphology and structure of transplanted ovarian tissues, facilitating graft function recovery, and extending lifespan. The amassed evidence supports the consideration of melatonin as a prospective protective agent for human ovarian tissue transplantation in the future.
Collapse
Affiliation(s)
- Lingyun Xie
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, 111 Ning Guo Road, Yangpu District, Shanghai 200011, China
| | - Yan Ding
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 128 Shen Yang Road, Yangpu District, Shanghai 200090, China
| | - Xuyin Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 128 Shen Yang Road, Yangpu District, Shanghai 200090, China.
| |
Collapse
|
24
|
Yang H, Lei G, Deng Z, Sun F, Tian Y, Cheng J, Yu H, Li C, Bai C, Zhang S, An G, Yang P. An Engineered Influenza a Virus Expressing the Co-Stimulator OX40L as an Oncolytic Agent Against Hepatocellular Carcinoma. J Hepatocell Carcinoma 2024; 11:1-13. [PMID: 38223555 PMCID: PMC10787515 DOI: 10.2147/jhc.s410703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 10/24/2023] [Indexed: 01/16/2024] Open
Abstract
Background Oncolytic virus (OV) therapy has emerged as a promising novel form of immunotherapy. Moreover, an increasing number of studies have shown that the therapeutic efficacy of OV can be further improved by arming OVs with immune-stimulating molecules. Methods In this study, we used reverse genetics to produce a novel influenza A virus, termed IAV-OX40L, which contained the immune-stimulating molecule OX40L gene in the influenza virus nonstructural (NS1) protein gene. The oncolytic effect of IAV-OX40L was explored on hepatocellular carcinoma (HCC)HCC cells in vitro and in vivo. Results Hemagglutination titers of the IAV-OX40L virus were stably 27-28 in specific-pathogen-free chicken embryos. The morphology and size distribution of IAV-OX40L are similar to those of the wild-type influenza. Expression of OX40L protein was confirmed by Western blot and immunofluorescence. MTS assays showed that the cytotoxicity of IAV-OX40L was higher in HCC cells (HepG2 and Huh7) than in normal liver cells (MIHA) in a time- and dose-dependent manner in vitro. We found that intratumoral injection of IAV-OX40L reduced tumor growth and increased the survival rate of mice compared with PR8-treated controls in vivo. In addition, the pathological results showed that IAV-OX40L selectively destroyed tumor tissues without harming liver and lung tissues. CD4+ and CD8+ T cells of the IAV-OX40L group were significantly increased in the splenic lymphocytes of mice. Further validation confirmed that IAV-OX40L enhanced the immune response mainly by activating Th1-dominant immune cells, releasing interferon-γ and interleukin-2. Conclusion Taken together, our findings demonstrate the novel chimeric influenza OV could provide a potential therapeutic strategy for combating HCC and improve the effectiveness of virotherapy for cancer therapy.
Collapse
Affiliation(s)
- Hao Yang
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- Department of Hepatological Surgery, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
- Department of Surgery, Taian City Central Hospital, Taian, People's Republic of China
| | - Guanglin Lei
- Department of Hepatological Surgery, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhuoya Deng
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Fang Sun
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Yuying Tian
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jinxia Cheng
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Hongyu Yu
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Cong Li
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Changqing Bai
- Department of Respiratory, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Guangdong, People's Republic of China
| | - Shaogeng Zhang
- Department of Hepatological Surgery, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Guangwen An
- Department of Pharmacy, No. 984 Hospital of the PLA, Beijing, People's Republic of China
| | - Penghui Yang
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
25
|
Martin PJ. CD24Fc to DAMPen GVHD. Blood 2024; 143:1-2. [PMID: 38175676 DOI: 10.1182/blood.2023022228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024] Open
Affiliation(s)
- Paul J Martin
- Fred Hutchinson Cancer Center and University of Washington
| |
Collapse
|
26
|
Magenau J, Jaglowski S, Uberti J, Farag SS, Riwes MM, Pawarode A, Anand S, Ghosh M, Maciejewski J, Braun T, Devenport M, Lu S, Banerjee B, DaSilva C, Devine S, Zhang MJ, Burns LJ, Liu Y, Zheng P, Reddy P. A phase 2 trial of CD24Fc for prevention of graft-versus-host disease. Blood 2024; 143:21-31. [PMID: 37647633 PMCID: PMC10934299 DOI: 10.1182/blood.2023020250] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/25/2023] [Accepted: 08/13/2023] [Indexed: 09/01/2023] Open
Abstract
ABSTRACT Patients who undergo human leukocyte antigen-matched unrelated donor (MUD) allogeneic hematopoietic stem cell transplantation (HSCT) with myeloablative conditioning for hematologic malignancies often develop acute graft-versus-host disease (GVHD) despite standard calcineurin inhibitor-based prophylaxis in combination with methotrexate. This trial evaluated a novel human CD24 fusion protein (CD24Fc/MK-7110) that selectively targets and mitigates inflammation due to damage-associated molecular patterns underlying acute GVHD while preserving protective immunity after myeloablative conditioning. This phase 2a, multicenter study evaluated the pharmacokinetics, safety, and efficacy of CD24Fc in combination with tacrolimus and methotrexate in preventing acute GVHD in adults undergoing MUD HSCT for hematologic malignancies. A double-blind, placebo-controlled, dose-escalation phase to identify a recommended dose was followed by an open-label expansion phase with matched controls to further evaluate the efficacy and safety of CD24Fc in preventing acute GVHD. A multidose regimen of CD24Fc produced sustained drug exposure with similar safety outcomes when compared with single-dose regimens. Grade 3 to 4 acute GVHD-free survival at day 180 was 96.2% (95% confidence interval [CI], 75.7-99.4) in the CD24Fc expansion cohort (CD24Fc multidose), compared with 73.6% (95% CI, 63.2-81.4) in matched controls (hazard ratio, 0.1 [95% CI, 0.0-0.6]; log-rank test, P = .03). No participants in the CD24Fc escalation or expansion phases experienced dose-limiting toxicities (DLTs). The multidose regimen of CD24Fc was well tolerated with no DLTs and was associated with high rates of severe acute GVHD-free survival after myeloablative MUD HSCT. This trial was registered at ClinicalTrials.gov as #NCT02663622.
Collapse
Affiliation(s)
- John Magenau
- Transplantation and Cellular Therapy Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Samantha Jaglowski
- The James Cancer Hospital and Solove Research Institute, Ohio State University, Columbus, OH
| | - Joseph Uberti
- Karmanos Cancer Center, Hudson-Webber Cancer Research Center, Detroit, MI
| | - Sherif S. Farag
- Blood and Bone Marrow Stem Cell Transplant and Immune Cell Therapy Program, Indiana University, Indianapolis, IN
| | - Mary Mansour Riwes
- Transplantation and Cellular Therapy Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Attaphol Pawarode
- Transplantation and Cellular Therapy Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Sarah Anand
- Transplantation and Cellular Therapy Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Monalisa Ghosh
- Transplantation and Cellular Therapy Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - John Maciejewski
- Transplantation and Cellular Therapy Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Thomas Braun
- Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI
| | | | | | | | | | | | - Mei-Jie Zhang
- Center for International Blood and Marrow Transplant Research, Milwaukee, WI
| | - Linda J. Burns
- Center for International Blood and Marrow Transplant Research, Milwaukee, WI
| | - Yang Liu
- OncoImmune, Inc, Rockville, MD
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD
| | - Pan Zheng
- OncoImmune, Inc, Rockville, MD
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD
| | - Pavan Reddy
- Transplantation and Cellular Therapy Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
27
|
Xu Y, Huang X, Nie XC, Liu YS, Zhou Y, Niu JM. Manganese and IL-12 treatment alters the ovarian tumor microenvironment. Aging (Albany NY) 2024; 16:191-206. [PMID: 38175694 PMCID: PMC10817382 DOI: 10.18632/aging.205361] [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: 08/06/2023] [Accepted: 11/06/2023] [Indexed: 01/05/2024]
Abstract
Metal immunotherapy is a novel adjuvant immunotherapy. Mn2+ can activate STING-a type I IFN response protein-that promotes innate immunity and increases anti-tumor activity by promoting macrophage phagocytosis. IL-12, a cytokine that increases the antigen-presenting ability to promote effector T-cell activation, has potent antitumor activity, albeit with severe adverse effects. In this study, we observed that the combination of Mn2+ and IL-12 has a better antitumor effect and possibly reflects a better safety profile, providing a novel approach and theoretical basis for safe and rapid cancer treatment.
Collapse
Affiliation(s)
- Yan Xu
- Department of Gynecology, Shenyang Women and Children’s Hospital, Shenyang 110000, China
| | - Xin Huang
- Department of General Practice Medicine, Shengjing Hospital Affiliated to China Medical University, Shenyang 110000, China
| | - Xiao-Cui Nie
- Department of Gynecology, Shenyang Women and Children’s Hospital, Shenyang 110000, China
| | - Yan-Song Liu
- Department of Gynecology, Shenyang Women and Children’s Hospital, Shenyang 110000, China
| | - Yang Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang 110000, China
| | - Ju-Min Niu
- Department of Gynecology, Shenyang Women and Children’s Hospital, Shenyang 110000, China
| |
Collapse
|
28
|
Brami I, Zuckerman T, Ram R, Avni B, Peretz G, Ostrovsky D, Lior Y, Faour C, McElvaney O, McElvaney NG, Lewis EC. Altered Serum Alpha1-Antitrypsin Protease Inhibition before and after Clinical Hematopoietic Stem Cell Transplantation: Association with Risk for Non-Relapse Mortality. Int J Mol Sci 2023; 25:422. [PMID: 38203593 PMCID: PMC10779144 DOI: 10.3390/ijms25010422] [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: 11/21/2023] [Revised: 12/23/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
α1-Antitrypsin (AAT), an acute-phase reactant not unsimilar to C-reactive protein (CRP), is a serine protease inhibitor that harbors tissue-protective and immunomodulatory attributes. Its concentrations appropriately increase during conditions of extensive tissue injury, and it induces immune tolerance, in part, by inhibiting the enzymatic activity of the inflammatory serine protease, proteinase 3 (PR3). Typically administered to patients with genetic AAT deficiency, AAT treatment was recently shown to improve outcomes in patients with steroid-refractory graft-versus-host disease (GVHD). GVHD represents a grave outcome of allogeneic hematopoietic stem cell transplantation (HSCT), a potentially curative intervention for hematological diseases. The procedure requires radio/chemotherapy conditioning of the prospective marrow recipient, a cytotoxic process that causes vast tissue injury and, in some formats, interferes with liver production of AAT. To date, changes in the functional profile of AAT during allogeneic HSCT, and during the cytotoxic intervention that precedes HSCT, are unknown. The present study followed 53 patients scheduled for allogeneic HSCT (trial registration NCT03188601). Serum samples were tested before and after HSCT for AAT and CRP levels and for intrinsic anti-proteolytic activity. The ex vivo response to clinical-grade AAT was tested on circulating patient leukocytes and on a human epithelial cell line treated with patient sera in a gap closure assay. According to the ex vivo experiments, circulating leukocytes responded to AAT with a favorable immune-regulated profile, and epithelial gap closure was enhanced by AAT in sera from GVHD-free patients but not in sera from patients who developed GVHD. According to serum collected prior to HSCT, non-relapse mortality was reliably predicted by combining three components: AAT and CRP levels and serum anti-proteolytic activity. Taken together, HSCT outcomes are significantly affected by the anti-proteolytic function of circulating AAT, supporting early AAT augmentation therapy for allogeneic HSCT patients.
Collapse
Affiliation(s)
- Ido Brami
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Be’er-Sheva 8410501, Israel;
| | - Tsila Zuckerman
- Hematology Department and Bone Marrow Transplantation Unit, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Ron Ram
- Bone Marrow Transplantation Unit, The Division of Hematology, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel;
| | - Batia Avni
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah-Hebrew University Medical Center, Ein Kerem, Jerusalem 9112001, Israel;
| | - Galit Peretz
- Department of Hematology, Soroka University Medical Center, Be’er-Sheva 8410101, Israel;
| | - Daniel Ostrovsky
- Clinical Research Center, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be’er-Sheva 8410101, Israel;
| | - Yotam Lior
- Division of Anesthesiology, Pain and Intensive Care, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel;
| | - Caroline Faour
- Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israeli Institute of Technology, Haifa 3109601, Israel;
| | - Oisin McElvaney
- The Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, D02 YN77 Dublin, Ireland; (O.M.); (N.G.M.)
| | - Noel G. McElvaney
- The Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, D02 YN77 Dublin, Ireland; (O.M.); (N.G.M.)
| | - Eli C. Lewis
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Be’er-Sheva 8410501, Israel;
| |
Collapse
|
29
|
Xu M, Cheng Y, Meng R, Yang P, Chen J, Qiao Z, Wu J, Qian K, Li Y, Wang P, Zhou L, Wang T, Sheng D, Zhang Q. Enhancement of Microglia Functions by Developed Nano-Immuno-Synergist to Ameliorate Immunodeficiency for Malignant Glioma Treatment. Adv Healthc Mater 2023; 12:e2301861. [PMID: 37573475 DOI: 10.1002/adhm.202301861] [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: 06/12/2023] [Revised: 08/02/2023] [Indexed: 08/14/2023]
Abstract
Resident microglia are key factors in mediating immunity against brain tumors, but the microglia in malignant glioma are functionally impaired. Little immunotherapy is explored to restore microglial function against glioma. Herein, oleanolic acid (OA) (microglia "restorer") and D PPA-1 peptide (immune checkpoint blockade) are integrated on a nano-immuno-synergist (D PAM@OA) to work coordinately. The self-assembled OA core is coated with macrophage membrane for efficient blood-brain barrier penetration and microglia targeting, on which D PPA-1 peptide is attached via acid-sensitive bonds for specific release in tumor microenvironment. With the enhanced accumulation of the dual drugs in their respective action sites, D PAM@OA effectively promotes the recruitment and activation of effector T cells by inhibiting aberrant activation of Signal transducer and activator of transcription (STAT-3) pathway in microglia, and assists activated effector T cells in killing tumor cells by blocking elevated immune checkpoint proteins in malignant glioma. Eventually, as adjuvant therapy, the rationally designed nano-immuno-synergist hinders malignant glioma progression and recurrence with or without temozolomide. The work demonstrates the feasibility of a nano-formulation for microglia-based immunotherapy, which may provide a new direction for the treatment of brain tumors.
Collapse
Affiliation(s)
- Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Ran Meng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Jian Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Zhen Qiao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Pengzhen Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Tianying Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Dongyu Sheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| |
Collapse
|
30
|
Qi L, Peng J, Huang X, Zhou T, Tan G, Li F. Longitudinal dynamics of gut microbiota in the pathogenesis of acute graft-versus-host disease. Cancer Med 2023; 12:21567-21578. [PMID: 38053512 PMCID: PMC10757094 DOI: 10.1002/cam4.6557] [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: 06/09/2023] [Revised: 08/27/2023] [Accepted: 09/09/2023] [Indexed: 12/07/2023] Open
Abstract
AIM The gut microbiota has been reported to be associated with acute graft-versus-host disease (aGvHD) in hematopoietic stem cell transplantation (HSCT). Dynamic surveillance of the microbiota is required to understand the detailed pathogenesis involved in the process of aGvHD. METHODS Fecal samples were collected prospectively at four timepoints, including pre-HSCT (T1), graft infusion (T2), neutrophil engraftment (T3), and 30 days after transplantation (T4). Fecal samples were profiled by 16S ribosomal RNA gene sequencing to assess the microbiota composition. RESULTS From the T1 to T4 timepoint, the diversity of the gut microbiota decreased, and the dominant species also changed, with a decrease in the obligate anaerobic bacteria and a shift toward a "pathogenic community". Compared with non-aGvHD patients, aGvHD patients had a lower abundance of Roseburia at T1 and a higher abundance of Acinetobacter johnsonii at T2. Furthermore, Acinetobacter johnsonii was negatively correlated with the secretion of IL-4 and TNF-α. At T3, Rothia mucilaginos was demonstrated to be linked with a decreased risk of aGvHD, which was accompanied by decreased secretion of IL-8. At T4, higher abundances of Lactobacillus paracasei and Acinetobacter johnsonii were identified to be related with aGvHD. Lactobacillus paracasei was associated with the downregulation of IL-10, and Acinetobacter johnsonii was associated with the downregulation of IL-2 and TNF-α. CONCLUSIONS Dynamic changes in gut microbiota composition and related cytokines were found to be related to aGvHD, including pathogenic or protective changes. These findings suggested that manipulation of gut microbiota at different timepoints might be a promising avenue for preventing or treating this common complication.
Collapse
Affiliation(s)
- Ling Qi
- Center of HematologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Clinical Research Center for Hematologic DiseaseNanchangChina
- Institute of Lymphoma and MyelomaNanchang UniversityNanchangChina
| | - Jie Peng
- Center of HematologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Clinical Medical College of Nanchang UniversityNanchangChina
| | - Xianbao Huang
- Center of HematologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Clinical Research Center for Hematologic DiseaseNanchangChina
- Institute of Lymphoma and MyelomaNanchang UniversityNanchangChina
| | - Ting Zhou
- Center of HematologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Clinical Research Center for Hematologic DiseaseNanchangChina
- Institute of Lymphoma and MyelomaNanchang UniversityNanchangChina
| | - Genmei Tan
- Center of HematologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Clinical Research Center for Hematologic DiseaseNanchangChina
- Institute of Lymphoma and MyelomaNanchang UniversityNanchangChina
| | - Fei Li
- Center of HematologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Clinical Research Center for Hematologic DiseaseNanchangChina
- Institute of Lymphoma and MyelomaNanchang UniversityNanchangChina
| |
Collapse
|
31
|
Zheng Q, Wang T, Jiang G, Li M, Zhang Z, Chen Y, Tian X. Immunoglobulin superfamily 6 is a molecule involved in the anti-tumor activity of macrophages in lung adenocarcinoma. BMC Cancer 2023; 23:1170. [PMID: 38037023 PMCID: PMC10688083 DOI: 10.1186/s12885-023-11681-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/27/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Immunoglobulin superfamily 6 (IGSF6) is a novel member of the immunoglobulin superfamily and has been implicated in various diseases. However, the specific role of IGSF6 in the anti-tumor immunity within lung adenocarcinoma (LUAD) remains unclear. METHODS We analyzed the IGSF6 expression in LUAD using data from TCGA, and we performed qRT-PCR and western blotting to validate these findings using tissue samples obtained from LUAD patients. Images of IHC staining were obtained from HPA. To assess the clinical relevance of IGSF6 expression, we utilized UALCAN and SPSS to analyze its association with major clinical features of LUAD. Additionally, we employed ROC curves and survival analysis to evaluate the potential diagnostic and prognostic value of IGSF6 in LUAD. To gain insights into the functional implications of IGSF6, we performed enrichment analysis using the R software clusterProfiler package. Moreover, we utilized TIMER2.0 and TISIDB to investigate the relationship between IGSF6 and immune infiltrates in LUAD. The proportion of tumor-infiltrating immune cells in LUAD was assessed using FCM, and their correlation with IGSF6 expression in tumor tissues was analyzed. The localization of IGSF6 protein on macrophages was confirmed using the HPA and FCM. To determine the regulatory role of IGSF6 on macrophage activity in LUAD, we employed ELISA, FCM, and tumor-bearing models. RESULTS We discovered that both IGSF6 mRNA and protein levels were significantly decreased in LUAD. Additionally, we observed a negative correlation between IGSF6 expression and TNM stages as well as pathologic stages in LUAD. Notably, IGSF6 exhibited high sensitivity and specificity in diagnosing LUAD, and was positively associated with the survival rate of LUAD patients. Furthermore, IGSF6 expression was closely linked to gene sets involved in immune response. IGSF6 expression showed a positive correlation with immune infiltrates exhibiting anti-tumor activity, particularly M1 macrophages. We confirmed the predominant localization of the IGSF6 protein on the membrane of M1 macrophages. Importantly, the knockdown of IGSF6 resulted in a reduction in the anti-tumor activity of M1 macrophages, thereby promoting tumor progression. CONCLUSION IGSF6 is a molecule that is essential for the anti-tumor activity of macrophages in LUAD.
Collapse
Affiliation(s)
- Qisi Zheng
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Ting Wang
- Department of Laboratory Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Gechen Jiang
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Miao Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Zhi Zhang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| | - Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.
| | - Xinyu Tian
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
32
|
Koyama M. A potential tissue-based biomarker in gut GVHD. Blood 2023; 142:1768-1769. [PMID: 37995106 DOI: 10.1182/blood.2023022229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2023] Open
|
33
|
Enriquez J, McDaniel Mims B, Stroever S, dos Santos AP, Jones-Hall Y, Furr KL, Grisham MB. Influence of Housing Temperature and Genetic Diversity on Allogeneic T Cell-Induced Tissue Damage in Mice. PATHOPHYSIOLOGY 2023; 30:522-547. [PMID: 37987308 PMCID: PMC10661280 DOI: 10.3390/pathophysiology30040039] [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: 09/22/2023] [Revised: 11/12/2023] [Accepted: 11/18/2023] [Indexed: 11/22/2023] Open
Abstract
The objective of this study was to determine how housing temperature and genetic diversity affect the onset and severity of allogeneic T cell-induced tissue damage in mice subjected to reduced intensity conditioning (RIC). We found that adoptive transfer of allogeneic CD4+ T cells from inbred donors into sub-lethally irradiated inbred recipients (I→I) housed at standard housing temperatures (ST; 22-24 °C) induced extensive BM and spleen damage in the absence of injury to any other tissue. Although engraftment of T cells in RIC-treated mice housed at their thermo-neutral temperature (TNT; 30-32 °C) also developed similar BM and spleen damage, their survival was markedly and significantly increased when compared to their ST counterparts. In contrast, the adoptive transfer of allogeneic T cells into RIC-treated outbred CD1 recipients failed to induce disease in any tissue at ST or TNT. The lack of tissue damage was not due to defects in donor T cell trafficking to BM or spleen but was associated with the presence of large numbers of B cells and myeloid cells within these tissues that are known to contain immunosuppressive regulatory B cells and myeloid-derived suppressor cells. These data demonstrate, for the first time, that housing temperature affects the survival of RIC-treated I→I mice and that RIC-conditioned outbred mice are resistant to allogeneic T cell-induced BM and spleen damage.
Collapse
Affiliation(s)
- Josue Enriquez
- Department of Microbiology and Immunology, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Brianyell McDaniel Mims
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Stephanie Stroever
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Andrea Pires dos Santos
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Yava Jones-Hall
- Department of Veterinary Pathobiology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Kathryn L. Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Matthew B. Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
34
|
Taylor JV, Callery EL, Rowbottom A. Optimisation of SARS-CoV-2 peptide stimulation and measurement of cytokine output by intracellular flow cytometry and bio-plex analysis. J Immunol Methods 2023; 522:113556. [PMID: 37683822 DOI: 10.1016/j.jim.2023.113556] [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: 07/21/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
Our study was conducted to optimise a peptide stimulation and an intracellular cytokine staining protocol, alongside Bio-Plex supernatant analysis, for use in patients who had previously contracted SARS-CoV-2 or received vaccination against this virus in a clinical laboratory setting. Peripheral Blood Mononuclear Cell extraction and cryopreservation allowed for cells to be stored long term and enhanced logistical processing of samples. Viability and functionality of cells were analysed by flow cytometric methodology using viability staining monoclonal antibodies conjugated to fluorochromes. Antibiotics and Benzonase Nuclease did not impact lymphocyte viability and so cell culture conditions were optimised in terms of retaining viability and functionality. Optimisation of peptide stimulation with Influenza and SARS-CoV-2 peptide pools was conducted through stimulation experiments assessing peptide concentration, peptide stimulation time and enrichment studies to increase precursor frequency. Cytokine output was measured by flow cytometry and Bio-Plex methodologies, with positive cytokine readings predominantly detected in the cell culture supernatant. Analysis of both intracellular and extracellular compartments allowed for detection of cytokines and established the retained cellular functionality post cryopreservation. These results also indicated that our peptide stimulation method can generate antigen-specific T lymphocytes upon exposure to SARS-CoV-2 peptide pools. Moreover, the measurement of specific cytokines could be applied to an array of conditions, such as chronic inflammatory diseases, but to also offer an alternative method of measuring vaccine responses. This platform is easily adaptable and can remain relevant alongside changing vaccine composition, thus ensuring its applicability to future vaccination programmes.
Collapse
Affiliation(s)
| | | | - Anthony Rowbottom
- Immunology Department at Lancashire Teaching Hospitals, United Kingdom
| |
Collapse
|
35
|
Jamy O, Zeiser R, Chen YB. Novel developments in the prophylaxis and treatment of acute GVHD. Blood 2023; 142:1037-1046. [PMID: 37471585 DOI: 10.1182/blood.2023020073] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/27/2023] [Accepted: 07/13/2023] [Indexed: 07/22/2023] Open
Abstract
Acute graft-versus-host disease (aGVHD) is a major life-threatening complication after allogeneic hematopoietic cell transplant. Traditional standard prophylaxis for aGVHD has included a calcineurin inhibitor plus an antimetabolite, whereas treatment has relied mainly on corticosteroids, followed by multiple nonstandard second-line options. In the past decade, this basic framework has been reshaped by approval of antithymocyte globulin products, the emergence of posttransplant cyclophosphamide, and recent pivotal trials studying abatacept and vedolizumab for GVHD prophylaxis, whereas ruxolitinib was approved for corticosteroid-refractory aGVHD treatment. Because of this progress, routine acute GVHD prophylaxis and treatment practices are starting to shift, and results of ongoing trials are eagerly awaited. Here, we review recent developments in aGVHD prevention and therapy, along with ongoing and future planned clinical trials in this space, outlining what future goals should be and the limitations of current clinical trial designs and end points.
Collapse
Affiliation(s)
- Omer Jamy
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Robert Zeiser
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany
| | - Yi-Bin Chen
- Hematopoietic Cell Transplant and Cell Therapy Program, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
36
|
Elhage A, Cuthbertson P, Sligar C, Watson D, Sluyter R. A Species-Specific Anti-Human P2X7 Monoclonal Antibody Reduces Graft-versus-Host Disease in Humanised Mice. Pharmaceutics 2023; 15:2263. [PMID: 37765233 PMCID: PMC10536354 DOI: 10.3390/pharmaceutics15092263] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/26/2023] [Accepted: 08/27/2023] [Indexed: 09/29/2023] Open
Abstract
Graft-versus-host disease (GVHD) is a T cell-mediated inflammatory disorder that arises from allogeneic haematopoietic stem cell transplantation and is often fatal. The P2X7 receptor is an extracellular adenosine 5'-triphosphate-gated cation channel expressed on immune cells. Blockade of this receptor with small molecule inhibitors impairs GVHD in a humanised mouse model. A species-specific blocking monoclonal antibody (mAb) (clone L4) for human P2X7 is available, affording the opportunity to determine whether donor (human) P2X7 contributes to the development of GVHD in humanised mice. Using flow cytometric assays of human RPMI 8266 and murine J774 cells, this study confirmed that this mAb bound and impaired human P2X7. Furthermore, this mAb prevented the loss of human regulatory T cells (hTregs) and natural killer (hNK) T cells in vitro. NOD-scid IL2Rγnull mice were injected with 10 × 106 human peripheral blood mononuclear cells (Day 0) and an anti-hP2X7 or control mAb (100 μg i.p. per mouse, Days 0, 2, 4, 6, and 8). The anti-hP2X7 mAb increased hTregs and hNK cells at Day 21. Moreover, anti-hP2X7 mAb-treatment reduced clinical and histological GVHD in the liver and lung compared to the control treatment at disease endpoint. hTregs, hNK, and hNK T cell proportions were increased, and human T helper 17 cell proportions were decreased at endpoint. These studies indicate that blockade of human (donor) P2X7 reduces GVHD development in humanised mice, providing the first direct evidence of a role for donor P2X7 in GVHD.
Collapse
Affiliation(s)
- Amal Elhage
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (A.E.); (P.C.); (C.S.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Peter Cuthbertson
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (A.E.); (P.C.); (C.S.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Chloe Sligar
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (A.E.); (P.C.); (C.S.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Debbie Watson
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (A.E.); (P.C.); (C.S.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Ronald Sluyter
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia; (A.E.); (P.C.); (C.S.); (D.W.)
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| |
Collapse
|
37
|
Bu X, Pan W, Wang J, Liu L, Yin Z, Jin H, Liu Q, Zheng L, Sun H, Gao Y, Ping B. Therapeutic Effects of HLA-G5 Overexpressing hAMSCs on aGVHD After Allo-HSCT: Involving in the Gut Microbiota at the Intestinal Barrier. J Inflamm Res 2023; 16:3669-3685. [PMID: 37645691 PMCID: PMC10461746 DOI: 10.2147/jir.s420747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023] Open
Abstract
Background Acute graft-versus-host disease (aGVHD) initiated by intestinal barrier dysfunction and gut microbiota dysbiosis, remains one of the main obstacles for patients undergoing allogenic hematopoietic stem cell transplantation (allo-HSCT) to achieve good prognosis. Studies have suggested that mesenchymal stem cells (MSCs) can suppress immune responses and reduce inflammation, and human leukocyte antigen-G5 (HLA-G5) plays an important role in the immunomodulatory effects of MSCs, but very little is known about the potential mechanisms in aGVHD. Thus, we explored the effect of HLA-G5 on the immunosuppressive properties of human amnion MSCs (hAMSCs) and demonstrated its mechanism related to the gut microbiota at the intestinal barrier in aGVHD. Methods Patients undergoing allo-HSCT were enrolled to detect the levels of plasma-soluble HLA-G (sHLA-G) and regulatory T cells (Tregs). Humanized aGVHD mouse models were established and treated with hAMSCs or HLA-G5 overexpressing hAMSCs (ov-HLA-G5-hAMSCs) to explore the mechanism of HLA-G5 mediated immunosuppressive properties of hAMSCs and the effect of ov-HLA-G5-hAMSCs on the gut microbiota at the intestinal barrier in aGVHD. Results The plasma levels of sHLA-G on day +30 after allo-HSCT in aGVHD patients were lower than those in patients without aGVHD, and the sHLA-G levels were positively correlated with Tregs percentages. ov-HLA-G5-hAMSCs had the potential to inhibit the expansion of CD3+CD4+ T and CD3+CD8+ T cells and promote Tregs differentiation, suppress proinflammatory cytokine secretion but promote anti-inflammatory cytokines release. Besides, ov-HLA-G5-hAMSCs also could reverse the intestinal barrier dysfunction and gut microbiota dysbiosis in aGVHD. Conclusion We demonstrated that HLA-G might work with Tregs to create a regulatory network together to reduce the occurrence of aGVHD. HLA-G5 mediated hAMSCs to exert higher immunosuppressive properties in vivo and reverse the immune imbalance caused by T lymphocytes and cytokines. Furthermore, HLA-G5 overexpressing hAMSCs could restore gut microbiota and intestinal barriers, thereby ameliorating aGVHD.
Collapse
Affiliation(s)
- Xiaoyin Bu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Department of Hematology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Weifeng Pan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Junhui Wang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Liping Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Zhao Yin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Hua Jin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Haitao Sun
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, People’s Republic of China
| | - Ya Gao
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Baohong Ping
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Department of Hematology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| |
Collapse
|
38
|
Li Q, Wang X, Song Q, Yang S, Wu X, Yang D, Marié IJ, Qin H, Zheng M, Nasri U, Kong X, Wang B, Lizhar E, Cassady K, Tompkins J, Levy D, Martin PJ, Zhang X, Zeng D. Donor T cell STAT3 deficiency enables tissue PD-L1-dependent prevention of graft-versus-host disease while preserving graft-versus-leukemia activity. J Clin Invest 2023; 133:e165723. [PMID: 37526084 PMCID: PMC10378157 DOI: 10.1172/jci165723] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 06/02/2023] [Indexed: 08/02/2023] Open
Abstract
STAT3 deficiency (STAT3-/-) in donor T cells prevents graft-versus-host disease (GVHD), but the impact on graft-versus-leukemia (GVL) activity and mechanisms of GVHD prevention remains unclear. Here, using murine models of GVHD, we show that STAT3-/- donor T cells induced only mild reversible acute GVHD while preserving GVL effects against nonsusceptible acute lymphoblastic leukemia (ALL) cells in a donor T cell dose-dependent manner. GVHD prevention depended on programmed death ligand 1/programmed cell death protein 1 (PD-L1/PD-1) signaling. In GVHD target tissues, STAT3 deficiency amplified PD-L1/PD-1 inhibition of glutathione (GSH)/Myc pathways that regulate metabolic reprogramming in activated T cells, with decreased glycolytic and mitochondrial ATP production and increased mitochondrial ROS production and dysfunction, leading to tissue-specific deletion of host-reactive T cells and prevention of GVHD. Mitochondrial STAT3 deficiency alone did not reduce GSH expression or prevent GVHD. In lymphoid tissues, the lack of host-tissue PD-L1 interaction with PD-1 reduced the inhibition of the GSH/Myc pathway despite reduced GSH production caused by STAT3 deficiency and allowed donor T cell functions that mediate GVL activity. Therefore, STAT3 deficiency in donor T cells augments PD-1 signaling-mediated inhibition of GSH/Myc pathways and augments dysfunction of T cells in GVHD target tissues while sparing T cells in lymphoid tissues, leading to prevention of GVHD while preserving GVL effects.
Collapse
Affiliation(s)
- Qinjian Li
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute of City of Hope, Duarte, California, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute of City of Hope, Duarte, California, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Qingxiao Song
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute of City of Hope, Duarte, California, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California, USA
- Fujian Medical University Center of Translational Hematology, Fujian Institute of Hematology, and Fujian Medical University Union Hospital, Fuzhou, China
| | - Shijie Yang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute of City of Hope, Duarte, California, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Xiwei Wu
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Dongyun Yang
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Isabelle J Marié
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
| | - Hanjun Qin
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Moqian Zheng
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute of City of Hope, Duarte, California, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Ubaydah Nasri
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute of City of Hope, Duarte, California, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Xiaohui Kong
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute of City of Hope, Duarte, California, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Bixin Wang
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute of City of Hope, Duarte, California, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California, USA
- Fujian Medical University Center of Translational Hematology, Fujian Institute of Hematology, and Fujian Medical University Union Hospital, Fuzhou, China
| | - Elizabeth Lizhar
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Kaniel Cassady
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute of City of Hope, Duarte, California, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Josh Tompkins
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute of City of Hope, Duarte, California, USA
| | - David Levy
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
| | - Paul J Martin
- Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China
| | - Defu Zeng
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute of City of Hope, Duarte, California, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
39
|
Handelsman S, Overbey J, Chen K, Lee J, Haj D, Li Y. PD-L1's Role in Preventing Alloreactive T Cell Responses Following Hematopoietic and Organ Transplant. Cells 2023; 12:1609. [PMID: 37371079 DOI: 10.3390/cells12121609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Over the past decade, Programmed Death-Ligand 1 (PD-L1) has emerged as a prominent target for cancer immunotherapies. However, its potential as an immunosuppressive therapy has been limited. In this review, we present the immunological basis of graft rejection and graft-versus-host disease (GVHD), followed by a summary of biologically relevant molecular interactions of both PD-L1 and Programmed Cell Death Protein 1 (PD-1). Finally, we present a translational perspective on how PD-L1 can interrupt alloreactive-driven processes to increase immune tolerance. Unlike most current therapies that block PD-L1 and/or its interaction with PD-1, this review focuses on how upregulation or reversed sequestration of this ligand may reduce autoimmunity, ameliorate GVHD, and enhance graft survival following organ transplant.
Collapse
Affiliation(s)
- Shane Handelsman
- BioMedical Engineering, Department of Orthopaedic Surgery, Homer Stryker MD School of Medicine (WMed), Western Michigan University, Kalamazoo, MI 49007, USA
| | - Juliana Overbey
- BioMedical Engineering, Department of Orthopaedic Surgery, Homer Stryker MD School of Medicine (WMed), Western Michigan University, Kalamazoo, MI 49007, USA
| | - Kevin Chen
- BioMedical Engineering, Department of Orthopaedic Surgery, Homer Stryker MD School of Medicine (WMed), Western Michigan University, Kalamazoo, MI 49007, USA
| | - Justin Lee
- BioMedical Engineering, Department of Orthopaedic Surgery, Homer Stryker MD School of Medicine (WMed), Western Michigan University, Kalamazoo, MI 49007, USA
| | - Delour Haj
- BioMedical Engineering, Department of Orthopaedic Surgery, Homer Stryker MD School of Medicine (WMed), Western Michigan University, Kalamazoo, MI 49007, USA
| | - Yong Li
- BioMedical Engineering, Department of Orthopaedic Surgery, Homer Stryker MD School of Medicine (WMed), Western Michigan University, Kalamazoo, MI 49007, USA
| |
Collapse
|
40
|
Patel DA, Crain M, Pusic I, Schroeder MA. Acute Graft-versus-Host Disease: An Update on New Treatment Options. Drugs 2023:10.1007/s40265-023-01889-2. [PMID: 37247105 DOI: 10.1007/s40265-023-01889-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2023] [Indexed: 05/30/2023]
Abstract
Acute graft-versus-host disease (GVHD) occurs in approximately 50% of patients and remains a primary driver of non-relapse and transplant-related mortality. The best treatment remains prevention with either in vivo or ex vivo T-cell depletion, with multiple strategies used worldwide based on factors such as institution preference, ability to perform graft manipulation, and ongoing clinical trials. Predicting patients at high risk for developing severe acute GVHD based on clinical and biomarker-based criteria allows for escalation or potential de-escalation of therapy. Modern therapies for treatment of the disease include JAK/STAT pathway inhibitors, which are standard of care in the second-line setting and are being investigated for upfront management of non-severe risk based on biomarkers. Salvage therapies beyond the second-line remain suboptimal. In this review, we will focus on the most clinically used GVHD prevention and treatment strategies, including the accumulating data on JAK inhibitors in both settings.
Collapse
Affiliation(s)
- Dilan A Patel
- Section of BMT & Leukemia, Division of Oncology, Department of Medicine, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Mallory Crain
- Section of BMT & Leukemia, Division of Oncology, Department of Medicine, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Iskra Pusic
- Section of BMT & Leukemia, Division of Oncology, Department of Medicine, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Mark A Schroeder
- Section of BMT & Leukemia, Division of Oncology, Department of Medicine, Washington University in St Louis School of Medicine, St Louis, MO, USA.
| |
Collapse
|
41
|
Zavaro M, Dangot A, Bar-Lev TH, Amit O, Avivi I, Ram R, Aharon A. The Role of Extracellular Vesicles (EVs) in Chronic Graft vs. Host Disease, and the Potential Function of Placental Cell-Derived EVs as a Therapeutic Tool. Int J Mol Sci 2023; 24:ijms24098126. [PMID: 37175831 PMCID: PMC10179565 DOI: 10.3390/ijms24098126] [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: 03/20/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Chronic graft-versus-host disease (cGVHD) presents with dermal inflammation and fibrosis. We investigated the characteristics of extracellular vesicles (EVs) obtained from cGVHD patients, and their potential effects on human dermal fibroblast (NHDF) cells. The anti-inflammatory and anti-fibrotic effects of placental EVs were also explored given their known anti-inflammatory properties. Fourteen cGVHD patients' EVs contained higher levels of fibrosis-related proteins, TGFβ and α-smooth muscle actin (αSMA), compared to EVs from thirteen healthy subjects. The exposure of NHDF cells to the patients' EVs increased the NHDF cells' TGFβ and αSMA expressions. Placental EVs derived from placental-expanded cells (PLX) (Pluri Inc.) and human villous trophoblast (HVT) cells expressing the mesenchymal markers CD29, CD73, and CD105, penetrated into both the epidermal keratinocytes (HACATs) and NHDF cells. Stimulation of the HACAT cells with cytokine TNFα/INFγ (0.01-0.1 ng/µL) reduced cell proliferation, while the addition of placental EVs attenuated this effect, increasing and normalizing cell proliferation. The treatment of NHDF cells with a combination of TGFβ and placental HVT EVs reduced the stimulatory effects of TGFβ on αSMA production by over 40% (p = 0.0286). In summary, EVs from patients with cGVHD can serve as a biomarker for the cGVHD state. Placental EVs may be used to regulate dermal inflammation and fibrosis, warranting further investigation of their therapeutic potential.
Collapse
Affiliation(s)
- Mor Zavaro
- Hematology Research Laboratory, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6195001, Israel
| | - Ayelet Dangot
- Hematology Research Laboratory, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6195001, Israel
| | - Tali Hana Bar-Lev
- Hematology Research Laboratory, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Odelia Amit
- The BMT Unit, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Irit Avivi
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6195001, Israel
- Hematology Department, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Ron Ram
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6195001, Israel
- The BMT Unit, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Anat Aharon
- Hematology Research Laboratory, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6195001, Israel
| |
Collapse
|
42
|
Zhang D, Liu Y, Ma J, Xu Z, Duan C, Wang Y, Li X, Han J, Zhuang R. Competitive binding of CD226/TIGIT with PVR regulates macrophage polarization and is involved in vascularized skin graft rejection. Am J Transplant 2023:S1600-6135(23)00404-5. [PMID: 37054890 DOI: 10.1016/j.ajt.2023.04.007] [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: 02/27/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 04/15/2023]
Abstract
End-stage organ failure often requires solid organ transplantation. Nevertheless, transplant rejection remains an unresolved issue. The induction of donor-specific tolerance is the ultimate goal in transplantation research. Here, an allograft vascularized skin rejection model using BALB/c-C57/BL6 mice was established to evaluate the regulation of the poliovirus receptor signaling pathway via CD226 knockout (KO) or TIGIT-Fc recombinant protein treatment. In the TIGIT-Fc-treated and CD226KO groups, graft survival time was significantly prolonged, with a Treg cell proportion increase and M2-type macrophage polarization. Donor-reactive recipient T cells became hyporesponsive while responding normally after a third-party antigen challenge. In both groups, serum IL-1β, IL-6, IL-12p70, IL-17A, TNF-α, IFN-γ, and monocyte chemoattractant protein-1 levels decreased, and the IL-10 level increased. In vitro, M2 markers, such as Arg1 and IL-10, were markedly increased by TIGIT-Fc, whereas iNOS, IL-1β, IL-6, IL-12p70, TNF-α, and IFN-γ levels decreased. CD226-Fc had the opposite effect. TIGIT suppressed Th1 and Th17 differentiation by inhibiting macrophage SHP-1 phosphorylation and enhanced ERK1/2-MSK1 phosphorylation and nuclear translocation of CREB. In conclusion, CD226 and TIGIT competitively bind to PVR with activating and inhibitory functions, respectively. Mechanistically, TIGIT promotes IL-10 transcription from macrophages by activating the ERK1/2-MSK1-CREB pathway and enhancing M2-type polarization. CD226/TIGIT-PVR are crucial regulatory molecules of allograft rejection.
Collapse
Affiliation(s)
- Dongliang Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yitian Liu
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jingchang Ma
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhigang Xu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Chujun Duan
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yuling Wang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xuemei Li
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Juntao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
43
|
Zhang J, Wang X, Wang R, Chen G, Wang J, Feng J, Li Y, Yu Z, Xiao H. Rapamycin Treatment Alleviates Chronic GVHD-Induced Lupus Nephritis in Mice by Recovering IL-2 Production and Regulatory T Cells While Inhibiting Effector T Cells Activation. Biomedicines 2023; 11:biomedicines11030949. [PMID: 36979928 PMCID: PMC10045991 DOI: 10.3390/biomedicines11030949] [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: 02/01/2023] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
In this study, we test the therapeutic effects of rapamycin in a murine model of SLE-like experimental lupus nephritis induced by chronic graft-versus-host disease (cGVHD). Our results suggest that rapamycin treatment reduced autoantibody production, inhibited T lymphocyte and subsequent B cell activation, and reduced inflammatory cytokine and chemokine production, thereby protecting renal function and alleviating histological lupus nephritis by reducing the occurrence of albuminuria. To explore the potential mechanism of rapamycin's reduction of kidney damage in mice with lupus nephritis, a series of functional assays were conducted. As expected, rapamycin remarkably inhibited the lymphocytes' proliferation within the morbid mice. Interestingly, significantly increased proportions of peripheral CD4+FOXP3+ and CD4+CD25high T cells were observed in rapamycin-treated group animals, suggesting an up-regulation of regulatory T cells (Tregs) in the periphery by rapamycin treatment. Furthermore, consistent with the results regarding changes in mRNA abundance in kidney by real-time PCR analysis, intracellular cytokine staining demonstrated that rapamycin treatment remarkably diminished the secretion of Th1 and Th2 cytokines, including IFN-γ, IL-4 and IL-10, in splenocytes of the morbid mice. However, the production of IL-2 from splenocytes in rapamycin-treated mice was significantly higher than in the cells from control group animals. These findings suggest that rapamycin treatment might alleviate systemic lupus erythematosus (SLE)-like experimental lupus nephritis through the recovery of IL-2 production, which promotes the expansion of regulatory T cells while inhibiting effector T cell activation. Our studies demonstrated that, unlike other commonly used immunosuppressants, rapamycin does not appear to interfere with tolerance induction but permits the expansion and suppressive function of Tregs in vivo.
Collapse
Affiliation(s)
- Jilu Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Department of Biomedicine, Institute of Frontier Medical Sciences, Jilin University, Changchun 130021, China
| | - Xun Wang
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Renxi Wang
- Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Ministry of Science and Technology, Beijing 100054, China
| | - Guojiang Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Jing Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yan Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zuyin Yu
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - He Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
44
|
|
45
|
Bastian D, Sui X, Choi HJ, Wu Y, Tian L, Yang K, Liu C, Liu Y, Yu XZ. The Absence of IL-12Rβ2 Expression on Recipient Nonhematopoietic Cells Diminishes Acute Graft-versus-Host Disease in the Gastrointestinal Tract. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:486-495. [PMID: 36548465 PMCID: PMC9938950 DOI: 10.4049/jimmunol.2200120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022]
Abstract
The gastrointestinal (GI) tract is a frequent target organ in acute graft-versus-host disease (aGVHD), which can determine the morbidity and nonrelapse mortality after allogeneic hematopoietic cell transplantation (allo-HCT). Donor T cells recognize allogeneic Ags presented by host APCs, proliferate, and differentiate into Th1 and Th17 cells that drive GVHD pathogenesis. IL-12 has been shown to play an important role in amplifying the allogeneic response in preclinical and clinical studies. This study demonstrates that IL-12Rβ2 expression on recipient nonhematopoietic cells is required for optimal development of aGVHD in murine models of allo-HCT. aGVHD attenuation by genetic depletion of IL-12R signaling is associated with reduced MHC class II expression by intestinal epithelial cells and maintenance of intestinal integrity. We verified IL-12Rβ2 expression on activated T cells and in the GI tract. This study, to our knowledge, reveals a novel function of IL-12Rβ2 in GVHD pathogenesis and suggests that selectively targeting IL-12Rβ2 on host nonhematopoietic cells may preserve the GI tract after allo-HCT.
Collapse
Affiliation(s)
- David Bastian
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Xiaohui Sui
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Hee-Jin Choi
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yongxia Wu
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Linlu Tian
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kaipo Yang
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Chen Liu
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Yuejun Liu
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Xue-Zhong Yu
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
- The Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
46
|
Abstract
When discovered in the early 2000s, interleukin-33 (IL-33) was characterized as a potent driver of type 2 immunity and implicated in parasite clearance, as well as asthma, allergy, and lung fibrosis. Yet research in other models has since revealed that IL-33 is a highly pleiotropic molecule with diverse functions. These activities are supported by elusive release mechanisms and diverse expression of the IL-33 receptor, STimulation 2 (ST2), on both immune and stromal cells. Interestingly, IL-33 also supports type 1 immune responses during viral and tumor immunity and after allogeneic hematopoietic stem cell transplantation. Yet the IL-33-ST2 axis is also critical to the establishment of systemic homeostasis and tissue repair and regeneration. Despite these recent findings, the mechanisms by which IL-33 governs the balance between immunity and homeostasis or can support both effective repair and pathogenic fibrosis are poorly understood. As such, ongoing research is trying to understand the potential reparative and regulatory versus pro-inflammatory and pro-fibrotic roles for IL-33 in transplantation. This review provides an overview of the emerging regenerative role of IL-33 in organ homeostasis and tissue repair as it relates to transplantation immunology. It also outlines the known impacts of IL-33 in commonly transplanted solid organs and covers the envisioned roles for IL-33 in ischemia-reperfusion injury, rejection, and tolerance. Finally, we give a comprehensive summary of its effects on different cell populations involved in these processes, including ST2 + regulatory T cells, innate lymphoid cell type 2, as well as significant myeloid cell populations.
Collapse
|
47
|
Zhao X, Wang W, Nie S, Geng L, Song K, Zhang X, Yao W, Qiang P, Sun G, Wang D, Liu H. Dynamic comparison of early immune reactions and immune cell reconstitution after umbilical cord blood transplantation and peripheral blood stem cell transplantation. Front Immunol 2023; 14:1084901. [PMID: 37114055 PMCID: PMC10126295 DOI: 10.3389/fimmu.2023.1084901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Umbilical cord blood transplantation (UCBT) and peripheral blood stem cell transplantation (PBSCT) are effective allogeneic treatments for patients with malignant and non-malignant refractory hematological diseases. However, the differences in the immune cell reconstitution and the immune reactions during initial stages post-transplantation are not well established between UCBT and PBSCT. Therefore, in this study, we analyzed the differences in the immune reactions during the early stages (days 7-100 post-transplantation) such as pre-engraftment syndrome (PES), engraftment syndrome (ES), and acute graft-versus-host disease (aGVHD) and the immune cell reconstitution between the UCBT and the PBSCT group of patients. We enrolled a cohort of patients that underwent UCBT or PBSCT and healthy controls (n=25 each) and evaluated their peripheral blood mononuclear cell (PBMC) samples and plasma cytokine (IL-10 and GM-CSF) levels using flow cytometry and ELISA, respectively. Our results showed that the incidences of early immune reactions such as PES, ES, and aGVHD were significantly higher in the UCBT group compared to the PBSCT group. Furthermore, in comparison with the PBSCT group, the UCBT group showed higher proportion and numbers of naïve CD4+ T cells, lower proportion and numbers of Tregs, higher proportion of CD8+ T cells with increased activity, and higher proportion of mature CD56dim CD16+ NK cells during the early stages post-transplantation. Moreover, the plasma levels of GM-CSF were significantly higher in the UCBT group compared to the PBSCT group in the third week after transplantation. Overall, our findings demonstrated significant differences in the post-transplantation immune cell reconstitution between the UCBT and the PBSCT group of patients. These characteristics were associated with significant differences between the UCBT and the PBSCT groups regarding the incidences of immune reactions during the early stages post transplantation.
Collapse
Affiliation(s)
- Xuxu Zhao
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wenya Wang
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shiqin Nie
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Liangquan Geng
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Kaidi Song
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xinyi Zhang
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wen Yao
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Ping Qiang
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Guangyu Sun
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Dongyao Wang
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui, China
- *Correspondence: Dongyao Wang, ; Huilan Liu,
| | - Huilan Liu
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Transfusion, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- *Correspondence: Dongyao Wang, ; Huilan Liu,
| |
Collapse
|
48
|
Zhao Y, Wang Z, Shi X, Liu T, Yu W, Ren X, Zhao H. Effect of Chemotherapeutics on In Vitro Immune Checkpoint Expression in Non-Small Cell Lung Cancer. Technol Cancer Res Treat 2023; 22:15330338231202307. [PMID: 37728201 PMCID: PMC10515539 DOI: 10.1177/15330338231202307] [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: 04/05/2023] [Revised: 07/03/2023] [Accepted: 07/28/2023] [Indexed: 09/21/2023] Open
Abstract
Objectives: Immune checkpoint (ICP) expression in tumor cells could directly or indirectly affect the results of immunotherapy. ICP ligands on tumor cells usually bind their immune cell receptors to inhibit the activity, resulting in tumor immune escape. Thus, the purpose of this study was to ascertain the impact of various chemotherapeutic drugs on ICP expression in non-small cell lung cancer (NSCLC) cell lines with different pathological subtypes to provide a basis for the development of a superior regimen of chemotherapy combined with ICP blockade. Methods: Several first-line chemotherapy agents (cisplatin, carboplatin, paclitaxel, gemcitabine, vinorelbine, and pemetrexed) were selected to treat different NSCLC cell lines (squamous carcinoma H1703, adenocarcinoma A549, and large cell cancer H460) for 72 hours, and then the changes in ICP expression in the tumor cells were observed through flow cytometry. Results: Cisplatin, carboplatin, and paclitaxel upregulated the expressions of programmed cell death ligand 1 (PD-L1) and programmed cell death ligand 2 (PD-L2) in A549 and H460 cell lines. Meanwhile, vinorelbine and pemetrexed upregulated PD-L1 and PD-L2 in H1703, A549, and H460 cell lines. Paclitaxel, gemcitabine, vinorelbine, and pemetrexed significantly upregulated the expressions of both galectin-9 and high-mobility group box protein 1 (HMGB1) in the A549 cell line. Cisplatin and paclitaxel significantly upregulated the expressions of major histocompatibility complex-II (MHC-II), galectin-3, α-synuclein, and fibrinogen-like protein 1 (FGL1) in A549 and H460 cell lines. In addition, cisplatin and vinorelbine significantly upregulated the expressions of both CD155 and CD112 in the H460 cell line. Vinorelbine upregulated MHC-I in all three cell lines. Conclusion: Chemotherapy agents have different effects on the expression of ICP ligands in tumor cells with different pathological types, and this may affect the efficacy of combined immunotherapy. These results provide a theoretical basis for further selection and optimization of the combination of chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Yu Zhao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Zhe Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiuhuan Shi
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Ting Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Wenwen Yu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Haihe Laboratory of Cell Ecosystem, Tianjin, China
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hua Zhao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Haihe Laboratory of Cell Ecosystem, Tianjin, China
| |
Collapse
|
49
|
Dos Santos Nunes Pereira AC, Chahin BM, Tarzia A, Vilela RM. Nutritional status and prognosis in children with immunodeficiencies undergoing hematopoietic stem cell transplantation. Clin Nutr ESPEN 2022; 52:1-11. [PMID: 36513439 DOI: 10.1016/j.clnesp.2022.09.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/29/2022] [Accepted: 09/26/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Primary immunodeficiencies (PID) are diseases resulting from genetic dysfunctions in the immune system, which can result in recurrent infections, autoimmunity and even malignancy. It is estimated that approximately one-third of the PID described have gastrointestinal components or symptoms involved and may present an increased risk of weight loss and failure to thrive. It is also known that, in patients with other diagnoses, malnutrition may be associated with worse outcomes after hematopoietic stem cell transplantation (HSCT). OBJECTIVE to characterize the nutritional status of pediatric patients with PID at the time of admission for HSCT and to establish the relationship between baseline nutritional status measures and post-HSCT clinical outcomes. METHODS a retrospective analytical observational study, based on data from pediatric patients, of both sexes and all ethnicities, with PID, submitted to HSCT in the period from 2004 to 2019. The risk factors analyzed were the Z-scores of weights for age (W/A), height for age (H/A), BMI for age (BMI/A) and Sum score, obtained by through the sum of the W/A and H/A scores. The primary outcomes were overall survival at 6 months, occurrence of acute Graft Versus Host Disease (aGVHD) at 6 months, and occurrence of chronic Graft Versus Host Disease (cGVHD) at 1 year. Secondary outcomes were occurrence and degree of mucositis, length of stay, and total number of infectious episodes. As statistical analysis, the ANOVA model, the Tukey test, ROC curves and Kaplan Meier and Log-Rank analysis were used. Multivariate survival and logistic regression models were also performed. RESULTS The study showed important indicators of malnutrition in patients with PID, especially those diagnosed with Severe Combined Immunodeficiency Syndrome (SCID) and Hemophagocytic Syndromes (HS). Among those with SCID, 60% had low or very low weight for their age, 52% had low or very short stature for their age, and 44% were classified as being thin or very thin. Among patients with HS, 75% had short or very short stature for their age. Multivariate analysis only demonstrated association between W/A score with extensive cGVHD, controlling for diagnosis, compatibility, conditioning and immunoprophylaxis. Lower W/A values were associated with higher occurrences of these events. Although W/A was only associated with cGVHD and H/A had no association with chronic or acute GVHD, when Sum scores were used, the lower values the higher rates of severe aGVHD and total cGVHD according to multivariate controlled models for diagnosis, compatibility, conditioning and immunoprophylaxis. CONCLUSIONS Our study characterized the nutritional status of children with PID undergoing HSCT and found alarming rates of underweight and short stature in patients with SCID and HS. We also demonstrated a relationship between anthropometric parameters and outcomes such as mortality, the occurrence of GVHD and severe mucositis after HSCT. In this sense, W/A and Sum score measures would be good prognostic methods for these outcomes. Henceforth, prospective studies are needed to confirm these findings and establish new nutritional assessment criteria for this population.
Collapse
Affiliation(s)
| | - Brenda Machado Chahin
- Resident Nutritionist of the Hospital Care Program in Oncology and Hematology of the Complex Hospital of Clinics UFPR, Brazil
| | - Andréa Tarzia
- Nutritionist of the Bone Marrow Transplant Service of the Complex Hospital of Clinics UFPR, Curitiba, PR, Brazil
| | - Regina Maria Vilela
- Professor at the Department of Nutrition of the Federal University of Paraná UFPR, Curitiba, PR, Brazil.
| |
Collapse
|
50
|
Cherk MH, Khor R, Barber TW, Yap KSK, Patil S, Walker P, Avery S, Roberts S, Kemp W, Pham A, Bailey M, Kalff V. Noninvasive Assessment of Acute Graft-Versus-Host Disease of the Gastrointestinal Tract After Allogeneic Hemopoietic Stem Cell Transplantation Using 18F-FDG PET. J Nucl Med 2022; 63:1899-1905. [PMID: 35450959 DOI: 10.2967/jnumed.121.263688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/08/2022] [Indexed: 01/11/2023] Open
Abstract
Acute graft-versus-host disease of the gastrointestinal tract (acute GIT-GVHD) often complicates allogeneic hemopoietic stem cell transplantation (AHSCT). 18F-FDG PET/CT is known to detect active inflammation and may be a useful noninvasive test for acute GIT-GVHD. The objective of this study was to evaluate the diagnostic utility of 18F-FDG PET/CT to noninvasively assess patients with clinically suspected acute GIT-GVHD. Fifty-one AHSCT patients with clinically suspected acute GIT-GVHD prospectively underwent 18F-FDG PET/CT scanning followed by upper and lower GIT endoscopy within 7 d. Endoscopic biopsies of 4 upper GIT and 4 colonic segments were obtained for histology to compare with corresponding quantitative segmental 18F-FDG PET/CT SUVmax Receiver-operating-characteristic curve (ROC) analysis was performed to determine predictive capacity of 18F-FDG PET/CT SUVmax for acute GIT-GVHD. A separate qualitative visual 18F-FDG PET/CT analysis was also performed for comparison. Results: Twenty-three of 51 (45.1%) patients had biopsy-confirmed acute GIT-GVHD, with 19 of 23 (82.6%) having upper GIT and 22 of 22 (100%) colonic involvement. One of 23 patients did not undergo a colonoscopy. GVHD involved the entire colon contiguously in 21 of 22 patients. For quantitative analysis, histology from 4 upper GIT and 4 colonic segments were compared with 18F-FDG PET/CT SUVmax Colonic segments positive for GVHD had a higher SUVmax (4.1 [95% CI, 3.6-4.5]) than did normal colonic segments (2.3 [1.9-2.7], P = 0.006). No difference was demonstrated in upper GIT segments. Quantitative 18F-FDG PET/CT yielded a 69% sensitivity, 57% specificity, 73% negative predictive value, and 59% positive predictive value for the detection of GVHD compared with 70%, 76%, 76%, and 68%, respectively, for qualitative analysis. Conclusion: 18F-FDG PET is a useful noninvasive diagnostic test for acute GIT-GVHD, which when present always involves the colon and usually in its entirety, suggesting colonic biopsy obtained by sigmoidoscopy is adequate for histologic confirmation when acute GIT-GVHD is suspected. Of note, 18F-FDG PET cannot distinguish acute GIT-GVHD from non-GVHD inflammatory changes in the colon.
Collapse
Affiliation(s)
- Martin H Cherk
- Department of Nuclear Medicine & PET, Alfred Hospital, Melbourne Australia; .,Monash University, Melbourne, Australia
| | - Robert Khor
- Department of Nuclear Medicine & PET, Alfred Hospital, Melbourne Australia
| | - Thomas W Barber
- Department of Nuclear Medicine & PET, Alfred Hospital, Melbourne Australia.,Monash University, Melbourne, Australia
| | - Kenneth S K Yap
- Department of Nuclear Medicine & PET, Alfred Hospital, Melbourne Australia.,Monash University, Melbourne, Australia
| | - Sushrut Patil
- Monash University, Melbourne, Australia.,Department of Haematology, Alfred Hospital, Melbourne, Australia
| | - Patricia Walker
- Department of Haematology, Alfred Hospital, Melbourne, Australia
| | - Sharon Avery
- Monash University, Melbourne, Australia.,Department of Haematology, Alfred Hospital, Melbourne, Australia
| | - Stuart Roberts
- Monash University, Melbourne, Australia.,Department of Gastroenterology, Alfred Hospital, Melbourne, Australia
| | - William Kemp
- Monash University, Melbourne, Australia.,Department of Gastroenterology, Alfred Hospital, Melbourne, Australia
| | - Alan Pham
- Monash University, Melbourne, Australia.,Department of Anatomical Pathology, Alfred Hospital, Melbourne, Australia; and
| | - Michael Bailey
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Victor Kalff
- Department of Nuclear Medicine & PET, Alfred Hospital, Melbourne Australia.,Monash University, Melbourne, Australia
| |
Collapse
|