1
|
King Z, Anosike BI, Offenbacher R, Strumph K. A 7-Year-Old Boy with Prolonged Fever. NEJM EVIDENCE 2025; 4:EVIDmr2400330. [PMID: 40261121 DOI: 10.1056/evidmr2400330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
AbstractMorning Report is a time-honored tradition in which physicians-in-training present cases to their colleagues and clinical experts to collaboratively explore a patient's presentation. The Morning Report section continues this tradition by presenting a patient's chief concern and clinical course, inviting readers to develop a differential diagnosis and uncover the final diagnosis alongside the authors. This report details the story of a previously healthy 7-year-old boy who presented with 10 days of fever after travel to Bangladesh. Through targeted history, physical examination, and diagnostic testing, an illness script is developed. As the case progresses, the differential diagnosis is refined until a final diagnosis is established.
Collapse
Affiliation(s)
- Zoe King
- from the Pediatric Hematology and Oncology and Pediatric Infectious Disease Fellowship Programs at The Children's Hospital at Montefiore, Albert Einstein College of Medicine
| | - Brenda I Anosike
- from the Pediatric Hematology and Oncology and Pediatric Infectious Disease Fellowship Programs at The Children's Hospital at Montefiore, Albert Einstein College of Medicine
| | - Rachel Offenbacher
- from the Pediatric Hematology and Oncology and Pediatric Infectious Disease Fellowship Programs at The Children's Hospital at Montefiore, Albert Einstein College of Medicine
| | - Kaitlin Strumph
- from the Pediatric Hematology and Oncology and Pediatric Infectious Disease Fellowship Programs at The Children's Hospital at Montefiore, Albert Einstein College of Medicine
| |
Collapse
|
2
|
Li L, Mo Y, Yu X, He B, Dai Y, Fan L, Yang S, Liu H. Causal relationship between immune cells, metabolites and polycystic ovary syndrome identified by Mendelian randomization and mediation analyses. Immunol Cell Biol 2025; 103:461-472. [PMID: 40135765 DOI: 10.1111/imcb.70016] [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: 09/02/2024] [Revised: 12/24/2024] [Accepted: 02/26/2025] [Indexed: 03/27/2025]
Abstract
Immune cells and blood metabolites play essential roles in the development of polycystic ovary syndrome (PCOS); however, it remains unclear whether blood metabolites mediate the causal relationship between immune cells and PCOS. This study aimed to delineate the causal relationships among immune cells, PCOS and potential blood metabolites through Mendelian randomization (MR). A two-sample MR analysis was conducted using inverse variance weighting as the primary method to determine the causation between immune cells and PCOS risk. This was supplemented by a two-step MR analysis to assess the mediating role of blood metabolites between immune cells and PCOS. In addition, a series of sensitivity analysis methods were employed to test the robustness of the results. We also performed a reverse MR to evaluate the possibility of reverse causal relationships. Our findings identified 22 immune cell phenotypes causally linked to PCOS, with 12 acting as risk factors and 10 as protective factors for PCOS. Furthermore, 45 blood metabolites or ratios were causally related to PCOS. Mediation analysis revealed that X-25519 levels mediated 9.2% of the causal relationship between the absolute count of CD28-CD25++ CD8br and PCOS. In addition, N-acetylglucosamine/n-acetylgalactosamine levels and adenosine 5'-monophosphate levels mediated 6.7% and -11.1%, respectively, in the causation between naive DN(CD4- CD8-) %T cell and PCOS. The aspartate-to-citrate ratio mediated 8.6% of the causal relationship between CD20- CD38- %B cells and PCOS. Finally, reverse MR studies did not identify any reverse causation between the 22 immune cell phenotypes and PCOS. This study elucidates the causal links between immune cells and PCOS, highlighting the potential roles of four blood metabolites in mediating the interaction between immune cells and PCOS, thus providing new targets for research and therapeutic interventions.
Collapse
Affiliation(s)
- Lan Li
- Gynecology Department, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- College of lntegrative Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Yang Mo
- College of lntegrative Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Ximing Yu
- College of lntegrative Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Bing He
- Gynecology Department, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yue Dai
- Gynecology Department, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Longlong Fan
- Gynecology Department, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Sijie Yang
- College of lntegrative Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Huiping Liu
- College of lntegrative Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
3
|
Xing J, Wang J, Han L, Wang Y, Sun X, Shi J, Kong Q, Sun K, Zhang B. Causal Association Between Inflammatory Proteins, Inflammatory Cells, and Cauda Equina Syndrome: A Two-Sample Mendelian Randomization. World Neurosurg 2025; 197:123826. [PMID: 40020998 DOI: 10.1016/j.wneu.2025.123826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Recent studies have shown that inflammation plays a crucial role in the progression of cauda equina syndrome (CES). However, the exact cause-and-effect relationship between them is still unclear. METHODS We used CES data from the FinnGen genome-wide association study (GWAS), containing 329 cases and 408,351 control patients. Inflammatory proteins data were obtained from a large scale GWAS of 14,828 European ancestry participants, and inflammatory cells data were obtained from a GWAS summary of 3757 Sardinians. We chose inverse variance weighted as the main method and the Cochrane Q test to assess heterogeneity in the results. The MR-Egger intercept test and MR pleiotropy residual sum and outliers test were used to evaluate the horizontal pleiotropy, and sensitivity analysis was performed by leave-one-out analysis. RESULTS We examined robust associations between inflammatory proteins, inflammatory cells, and CES using Mendelian randomization. Two inflammatory proteins and 12 inflammatory cells were found as risk factors for CES: IL-8 and PD-L1; and basophil plasmacytoid dendritic cell, CD86+plasmacytoid dendritic cell, CD62L-plasmacytoid dendritic cell, CD39+secreting Treg, IgD+CD38-B cell, switched memory B cell, IgD+CD24+B cell, CD62L+dendritic cell, CD4+T cell, γδ T cell, and CD33dim HLA DR-myeloid cell. Two inflammatory proteins and 7 inflammatory cells were found as protective factors for CES: IL-10RA and CCL25; and transitional B cell, terminal differentiation double negative T cell, CD28-CD127-CD25++CD8br T cell, IgD+CD38br B cell, CD28+CD45RA-CD8br Treg, IgD+CD38-naive B cell, and granulocyte. Heterogeneity and pleiotropy analysis confirmed the reliability of the results. Our study reveals the causal relationship between inflammatory proteins, inflammatory cells, and CES, offering new insights for the development of future therapeutic drugs and early warning indicators. CONCLUSIONS Our findings extend genetic research to causal analysis between inflammatory proteins, cells, and CES. We found 2 proteins and 12 cells as risk factors and 2 proteins and 7 cells as protective factors. Further investigations are needed to verify whether these inflammation markers can be used to prevent or treat CES.
Collapse
Affiliation(s)
- Jianpeng Xing
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jinyu Wang
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Linhui Han
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yuan Wang
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiaofei Sun
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jiangang Shi
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Qingjie Kong
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kaiqiang Sun
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Bin Zhang
- Department of Orthopedic Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
4
|
Chen Y, Yu X, Cai Y, Jin Z, Xu Y. Large-scale mediator Mendelian randomization analysis identifies multiple immune cells mediating the causal link between gut microbes and chronic graft-versus-host disease risk. Exp Hematol 2025; 147:104794. [PMID: 40316245 DOI: 10.1016/j.exphem.2025.104794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/03/2025] [Accepted: 04/20/2025] [Indexed: 05/04/2025]
Abstract
Disorders of gut microbiota and immune cells have been observed to be involved in the occurrence of chronic graft-versus-host disease (cGVHD), but their causal connections have yet to be fully understood. This study utilized Mendelian randomization (MR), integrating genome-wide association study (GWAS) meta-analyses from the MiBioGen consortium (microbial taxa), the SardiNIA project (immune traits), and disease data from the Fred Hutchinson Cancer Research Center (FHCRC) cohort to investigate their relationships. The aim was to explore the causal effects of microbiota and immune traits on the incidence of cGVHD, using mediation analysis to identify which immune traits might mediate the effects of microbiota on this condition. The main analysis observed significant causal associations of 3 specific microbial taxa with cGVHD: Lactococcus.id.1851 (odds ratio [OR] = 1.989, 95% confidence interval [CI] = 1.311-3.019, p = 0.001), Ruminiclostridium9.id.11357 (OR = 3.273, 95% CI = 1.604-6.679, p = 0.001), and Intestinimonas.id.2062 (OR = 0.400, 95% CI = 0.230-0.697, p = 0.001). Sensitivity analysis and multivariable MR analysis ruled out possible horizontal pleiotropy and bias. Additionally, 10 immune traits, predominantly covering regulatory T cells (Tregs) and B cells, were identified as influencing cGVHD risk. The two-step mediation MR analysis presented the effect of identified microbial taxa on Tregs and B cells and detailed the pathways through which Intestinimonas impacts cGVHD via CD27 on memory B cells (proportion mediated = 4.2%). Similarly, the role of interactions between Ruminiclostridium9 and effector memory double-negative T cells in mediating cGVHD was quantified, accounting for 9.5% of the total effect.
Collapse
Affiliation(s)
- Yiyin Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xinghao Yu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| | - Yiming Cai
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Zhou Jin
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China; Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| |
Collapse
|
5
|
Zhang C, Sun G, Jin H, Wei Y, Zheng S, Wang X, Zhao X, Zhang D, Jia J. Double-negative T cells in combination with ursodeoxycholic acid ameliorates immune-mediated cholangitis in mice. BMC Med 2025; 23:209. [PMID: 40189495 PMCID: PMC11974204 DOI: 10.1186/s12916-025-04043-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 03/27/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Primary biliary cholangitis (PBC) is a liver-specific autoimmune disease. Treatment of PBC with ursodeoxycholic acid (UDCA) is not sufficient to prevent disease progression. Our previous study revealed that the number of hepatic double-negative T cells (DNT), which are unique regulatory T cells, was reduced in PBC patients. However, whether replenishment of DNT can prevent the progression of PBC remains unclear. METHODS DnTGFβRII (Tg) mice and 2OA-BSA-immunized mice received DNT alone or in combination with oral UDCA. After 6-12 weeks of treatment, these mice were assessed for serological changes, liver pathological manifestations and intrahepatic immune responses. RESULTS Adoptive transfer of DNT alone significantly decreased serum levels of alanine transaminase (ALT), aspartate transaminase (AST), antimitochondrial antibody M2 (AMA-M2) and immunoglobulin M (IgM) in both Tg and 2OA-BSA-immunized PBC mouse models. In addition, DNT exhibited a strong killing effect on liver T cells and strong inhibition of their proliferation, but did not significantly improve the histology of PBC liver. However, combination therapy with DNT and oral UDCA predominantly ameliorated liver inflammation and significantly inhibited hepatic T and B cells. In vitro further study revealed that UDCA up-regulated the proliferation of DNT, increased the expression of the functional molecule perforin, and reduced the expression of NKG2A and endothelial cell protein C receptor (EPCR) through the farnesoid X receptor (FXR)/JNK signaling pathway in both mice and human DNT. CONCLUSIONS A single transfer of DNT ameliorated PBC in mice, while combination therapy of DNT with oral UDCA displayed a better efficacy, with stronger inhibition of hepatic T and B cells. This study highlights the potential application of DNT-based combination therapy for PBC, especially for UDCA non-responders.
Collapse
Affiliation(s)
- Chunpan Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing, 100050, China
| | - Guangyong Sun
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 South Gongti Road, Beijing, 100020, China
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Hua Jin
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 South Gongti Road, Beijing, 100020, China
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yunxiong Wei
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 South Gongti Road, Beijing, 100020, China
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Shimeng Zheng
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 South Gongti Road, Beijing, 100020, China
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xiyu Wang
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 South Gongti Road, Beijing, 100020, China
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xinyan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
- National Clinical Research Center for Digestive Disease, Beijing, 100050, China
| | - Dong Zhang
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 South Gongti Road, Beijing, 100020, China.
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
- Beijing Laboratory of Oral Health, Capital Medical University School of Basic Medicine, Beijing, 100069, China.
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
- National Clinical Research Center for Digestive Disease, Beijing, 100050, China.
| |
Collapse
|
6
|
Willison AG, Hagler R, Weise M, Elben S, Huntemann N, Masanneck L, Pfeuffer S, Lichtenberg S, Golombeck KS, Preuth LM, Rolfes L, Öztürk M, Ruck T, Melzer N, Korsen M, Hauser SL, Hartung HP, Lang PA, Pawlitzki M, Räuber S, Meuth SG. Effects of Anti-CD20 Antibody Therapy on Immune Cell Dynamics in Relapsing-Remitting Multiple Sclerosis. Cells 2025; 14:552. [PMID: 40214505 PMCID: PMC11988809 DOI: 10.3390/cells14070552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
INTRODUCTION The efficacy of anti-CD20 antibodies has significantly contributed to advancing our understanding of disease pathogenesis and improved treatment outcomes in relapsing-remitting multiple sclerosis (RRMS). A comprehensive analysis of the peripheral immune cell profile, combined with prospective clinical characterization, of RRMS patients treated with ocrelizumab (OCR) or ofatumumab (OFA) was performed to further understand immune reconstitution following B-cell depletion. METHODS REBELLION-MS is a longitudinal analysis of RRMS patients treated with either OCR (n = 34) or OFA (n = 25). Analysis of B, T, natural killer (NK) and natural killer T (NKT) cells at baseline, month 1, and 12 was performed by multidimensional flow cytometry. Data were analyzed by conventional gating and unsupervised computational approaches. In parallel, different clinical parameters were longitudinally assessed. Twenty treatment-naïve age/sex-matched RRMS patients were included as the control cohort. RESULTS B-cell depletion by OCR and OFA resulted in significant reductions in CD20+ T and B cells as well as B-cell subsets, alongside an expansion of CD5+CD19+CD20- B cells, while also elevating exhaustion markers (CTLA-4, PD-1, TIGIT, TIM-3) across T, B, NK, and NKT cells. Additionally, regulatory T-cell (TREG) numbers increased, especially in OCR-treated patients, and reductions in double-negative (CD3+CD4-CD8-) T cells (DN T cells) were observed, with these DN T cells having higher CD20 expression compared to CD4 or CD8 positive T cells. These immune profile changes correlated with clinical parameters, suggesting pathophysiological relevance in RRMS. CONCLUSIONS Our interim data add weight to the argumentation that the exhaustion/activation markers, notably TIGIT, may be relevant to the pathogenesis of MS. In addition, we identify a potentially interesting increase in the expression of CD5+ on B cells. Finally, we identified a population of double-negative T cells (KLRG1+HLADR+, in particular) that is associated with MS activity and decreased with CD20 depletion.
Collapse
Affiliation(s)
- Alice G. Willison
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Ramona Hagler
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Margit Weise
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Saskia Elben
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Niklas Huntemann
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Lars Masanneck
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Steffen Pfeuffer
- Department of Neurology, University Hospital Giessen and Marburg, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Stefanie Lichtenberg
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- Core Facility Flow Cytometry, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Kristin S. Golombeck
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Lara-Maria Preuth
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Leoni Rolfes
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Menekse Öztürk
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Tobias Ruck
- Department of Neurology with Heimer Institute for Muscle Research, University Hospital Bergmannsheil, 44789 Bochum, Germany
| | - Nico Melzer
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Melanie Korsen
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Stephen L. Hauser
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA 94107, USA
| | - Hans-Peter Hartung
- Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
- Brain and Mind Center, University of Sydney, Sydney, NSW 2050, Australia
- Department of Neurology, Palacky University, 771 46 Olomouc, Czech Republic
| | - Philipp A. Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Saskia Räuber
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Sven G. Meuth
- Department of Neurology, Medical Faculty and University Hospital, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
7
|
Wu J, Liang J, Li S, Lu J, Zhou J, Gao M, Zhang Y, Chen J. DNA nanovaccines derived from ferritin-modified glycogens for targeted delivery to immature dendritic cells and for promotion of Th1 cell differentiation. Acta Biomater 2025; 196:436-452. [PMID: 40023466 DOI: 10.1016/j.actbio.2025.02.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
DNA vaccines have emerged as a powerful approach for advanced cancer therapy. Despite the development of various delivery systems to enhance the immunogenicity of DNA vaccines, many still face challenges such as limited DNA condensation, rapid degradation in vivo and insufficient targeting to lymph nodes (LNs). Synthetic dendrimers with modifiable surfaces exhibit high efficiency in DNA condensation, but their synthesis is extremely complex. This study utilizes cationic glycogen, a natural branched dendrimer-like polymer, as the core structure for efficient DNA condensation and delivery, ensuring good biocompatibility. By connecting ferritin light chain to the glycogen surfaces, active targeting of LNs can be achieved due to its affinity for the SCARA5 receptor on immature dendritic cells (DCs), facilitating vaccine migration to the LNs. In addition, a seperate plasmid encoding adjuvant IL-12 was co-delivered to further boost the immunogenicity of the DNA nanovaccine. In vivo and in vitro experiments confirmed the effective transfection capability of this DNA vaccine, demonstrating promoted DC maturation, increased antigen presentation, and Th1 cell differentiation, resulting in improved anti-tumor efficiency in vivo. This innovative multi-gene co-loaded DNA vaccine offers valuable insights into combined gene therapy and broadens the research horizon on non-viral gene carriers. STATEMENT OF SIGNIFICANCE: The DNA vaccine encounters challenges such as limited DNA condensation, rapid degradation and insufficient targeting to lymph nodes (LNs), resulting in generally weak immunogenicity. In the current study, a novel nanovaccine is developed by connecting ferritin light chain to natural dendrimer glycogen, for simultaneous delivery of dual plasmids. The cationized glycogen provides strong DNA condensation ability, while ensuring excellent stability of the nanovaccine. The presence of ferritin light chain leads to effective targeting of dendritic cells (DCs), facilitating its migration to LNs. Moreover, the plasmid encoding the adjuvant IL-12 is co-incorporated with the antigen plasmid to mitigate the immunosuppression environment. This strategy significantly improves the immunogenicity of DNA vaccines, demonstrating high efficiency in cancer immunotherapy.
Collapse
Affiliation(s)
- Jun Wu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China; School of Chemical & Material Engineering, Jiangnan University, Wuxi 214122, PR China
| | - Jing Liang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Sichen Li
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Jinjin Lu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Juan Zhou
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Min Gao
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China
| | - Yan Zhang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China.
| | - Jinghua Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, PR China.
| |
Collapse
|
8
|
Burks J, Tiwary S, Stevens DM, Skoczen SL, Kularatne RN, Stern ST, Berzofsky JA. PLS-α-GalCer: a novel targeted glycolipid therapy for solid tumors. J Immunother Cancer 2025; 13:e009539. [PMID: 40121031 PMCID: PMC11931891 DOI: 10.1136/jitc-2024-009539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 02/16/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND The prototypical type I natural killer T (NKT) cell agonist, α-galactosylceramide (α-GalCer), has shown only minimal effects against solid tumors in the clinic. The most promising clinical application of α-GalCer currently entails ex vivo priming of patient-derived dendritic cells; however, this technology suffers from cost, logistical concerns, and safety issues. As a parenteral dendritic cell-targeted alternative, we demonstrate that poly(L-lysine succinylated) (PLS)-α-GalCer, a novel scavenger receptor-A1 targeted α-GalCer prodrug has enhanced antitumor activity compared with α-GalCer. METHODS To compare the antitumor activity of PLS-α-GalCer and α-GalCer, we used mouse syngeneic subcutaneous pancreatic and cervical tumor models using Panc02 and TC-1 cells, respectively. Intratumoral immune cell infiltration was evaluated using flow cytometry and immunohistochemistry whole-slide scan analysis. Serum cytokine levels were examined by ELISA and LEGENDplex analysis. Type I NKT cell intracellular interferon-gamma (IFN-γ) levels were determined by flow cytometry. Immunofluorescence was used to test the uptake and processing of PLS-α-GalCer and α-GalCer in antigen-presenting cells (APCs). RESULTS The scavenger receptor A1 (SR-A1)-mediated targeting of α-GalCer to APCs by PLS-α-GalCer significantly improves the antitumor function against solid tumors compared with α-GalCer. The Panc02 and TC-1 tumor models demonstrated that PLS-α-GalCer increases intratumoral antigen-specific T, NKT and T cells, and increases the M1/M2 macrophage ratio. In the TC-1 tumor model, we demonstrated that PLS-α-GalCer synergizes with an E7 tumor vaccine to significantly suppress tumor growth and increase the survival of mice. Furthermore, the antitumor function of PLS-α-GalCer is dependent on type I NKT cells and requires SR-A1 targeting. In addition, using SR-A1 knockout RAW cells, a murine macrophage cell line, we showed that PLS-α-GalCer uptake and processing in APCs are more efficient compared with α-GalCer. PLS-α-GalCer also induces significantly less serum Th2 and Th17 cytokines while stimulating significantly more IFN-γ for a longer period and increases Th1:Th2 cytokine ratios compared with α-GalCer. CONCLUSIONS PLS-α-GalCer is a promising immunotherapy for the treatment of solid tumors that has superior antitumor activity compared with α-GalCer and could be combined with tumor vaccines and potentially other immunotherapies such as immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Julian Burks
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Shweta Tiwary
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - David M Stevens
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Frederick, Maryland, USA
| | - Sarah L Skoczen
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Frederick, Maryland, USA
| | - Ruvanthi N Kularatne
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Frederick, Maryland, USA
| | - Stephan T Stern
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Frederick, Maryland, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
9
|
Fajardo-Despaigne JE, Lombard-Vadnais F, Pelletier AN, Olazabal A, Boutin L, Pasquin S, Janelle V, Legault L, Delisle JS, Hillhouse EE, Coderre L, Lesage S. Characterization and effective expansion of CD4 -CD8 - TCRαβ + T cells from individuals living with type 1 diabetes. Mol Ther Methods Clin Dev 2025; 33:101400. [PMID: 39877593 PMCID: PMC11772147 DOI: 10.1016/j.omtm.2024.101400] [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: 07/13/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025]
Abstract
CD4-CD8- TCRαβ+ (double-negative [DN]) T cells represent a rare T cell population that promotes immunological tolerance through various cytotoxic mechanisms. In mice, autologous transfer of DN T cells has shown protective effects against autoimmune diabetes and graft-versus-host disease. Here, we characterized human DN T cells from people living with type 1 diabetes (PWT1D) and healthy controls. We found that while DN T cells and CD8+ T cells share many similarities, DN T cells are a unique T cell population, both at the transcriptomic and protein levels. We also show that by using various cytokine combinations, human DN T cells can be expanded in vitro up to 1,000-fold (mean >250-fold) and remain functional post-expansion. In addition, we report that DN T cells from PWT1D display a phenotype comparable to that of healthy controls, efficiently expand, and are highly functional. As DN T cells are immunoregulatory and can prevent T1D in various mouse models, these observations suggest that autologous DN T cells may be amenable to therapy for the prevention or treatment of T1D.
Collapse
Affiliation(s)
| | - Félix Lombard-Vadnais
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | | | - Aïnhoa Olazabal
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Lucie Boutin
- Département de Recherche Clinique, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC, Canada
| | - Sarah Pasquin
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Valérie Janelle
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
| | - Laurent Legault
- Département de Recherche Clinique, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC, Canada
- Department of Pediatrics, Montreal Children’s Hospital, Montreal, QC, Canada
| | - Jean-Sébastien Delisle
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Erin E. Hillhouse
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Lise Coderre
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Sylvie Lesage
- Immunologie-Oncologie, Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
10
|
Shafer AM, Kenna E, Golden LAF, Elhossiny AM, Perry KD, Wilkowski J, Yan W, Kaczkofsky B, McGue J, Bresler SC, Courtney AH, Dalman JM, Galban CJ, Jiang W, Espinoza CE, Chugh R, Iyer MK, Frankel TL, Pasca di Magliano M, Dlugosz AA, Angeles CV. An immunocompetent mouse model of liposarcoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.634916. [PMID: 40297505 PMCID: PMC12036434 DOI: 10.1101/2025.01.31.634916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Liposarcoma (LPS) is the most prevalent soft tissue sarcoma. The most common biological subtypes are well-differentiated (WDLPS), a low-grade disease that can evolve to high-grade dedifferentiated LPS (DDLPS), with increased rates of recurrence and metastasis and low response rates to chemotherapy and targeted therapies. Preclinical testing of immunotherapeutics for LPS has been held back by the lack of an immunocompetent mouse model. Here, we present a spontaneous immunocompetent LPS mouse model, ACPP, with targeted deletion of Trp53 and Pten in adipocytes to mimic signaling alterations observed in human LPS. Similar to human LPS, tumors arising in ACPP mice produce WDLPS and DDLPS, along with tumors that exhibit both WD and DD components. Murine and human DDLPS tumors possess transcriptional similarities, including increased expression of oncogenes Cdk4 and Hmga2 and reduced expression of the tumor suppressor Cebpa; further, both mouse and human DDLPS exhibit either high or low T cell infiltration. Syngeneic cell lines derived from spontaneous ACPP DDLPS reliably produce tumors following orthotopic injection, each with distinct growth patterns, aggressiveness and tumor infiltrating lymphocyte profiles. These models provide much needed tools to understand the complex immunobiology of LPS and greatly accelerate the pace of preclinical studies to uncover new therapies for patients with this aggressive malignancy.
Collapse
|
11
|
Prosser AC, Klenerman P, Lucas M. Understanding Liver Transplantation Outcomes Through the Lens of Its Tissue-resident Immunobiome. Transplantation 2025:00007890-990000000-00973. [PMID: 39780303 DOI: 10.1097/tp.0000000000005303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Tissue-resident lymphocytes (TRLs) provide a front-line immunological defense mechanism uniquely placed to detect perturbations in tissue homeostasis. The heterogeneous TRL population spans the innate to adaptive immune continuum, with roles during normal physiology in homeostatic maintenance, tissue repair, pathogen detection, and rapid mounting of immune responses. TRLs are especially enriched in the liver, with every TRL subset represented, including liver-resident natural killer cells; tissue-resident memory B cells; conventional tissue-resident memory CD8, CD4, and regulatory T cells; and unconventional gamma-delta, natural killer, and mucosal-associated invariant T cells. The importance of donor- and recipient-derived TRLs after transplantation is becoming increasingly recognized, although it has not been examined in detail after liver transplantation. This review summarizes the evidence for the roles of TRLs in liver transplant immunology, focusing on their features, functions, and potential for their harnessing to improve transplant outcomes.
Collapse
Affiliation(s)
- Amy C Prosser
- Medical School, University of Western Australia, Perth, WA, Australia
| | - Paul Klenerman
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Michaela Lucas
- Medical School, University of Western Australia, Perth, WA, Australia
- Department of Immunology, PathWest Laboratory Medicine, Perth, WA, Australia
- Department of Immunology, Sir Charles Gairdner Hospital, Perth, WA, Australia
- Department of Immunology, Perth Children's Hospital, Perth, WA, Australia
| |
Collapse
|
12
|
Zhu HR, Wei YB, Guo JQ, Liu XF. Double-negative T cells with a distinct transcriptomic profile are abundant in the peripheral blood of patients with breast cancer. Breast Cancer Res Treat 2025; 209:103-115. [PMID: 39254769 PMCID: PMC11785702 DOI: 10.1007/s10549-024-07477-6] [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: 07/10/2024] [Accepted: 08/25/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Double-negative T (DNT) cells comprise a distinct subset of T lymphocytes that have been implicated in immune responses. The aim of this study was to characterize the peripheral DNT population in breast cancer (BC) patients. METHODS DNT cells were isolated from the peripheral blood samples of BC patients and healthy controls by flow cytometry. The sorted DNT cells were analyzed by the Smart-seq2 for single-cell full-length transcriptome profiling. The differentially expressed genes (DEGs) between the BC and control groups were screened and functionally annotated by Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses using R. The protein-protein interaction (PPI) network of the DEGs was constructed using the CytoHubba and MCODE plug-in of Cytoscape software to identify the core genes. Survival status, DNA methylation level, immune infiltration and immune checkpoint expression were analyzed using Kaplan-Meier Plotter, UALCAN, MethSeuvr, TIMER, and TISIDB respectively. The sequencing results were verified by RT-qPCR. RESULT The percentage of DNT cells was higher in the BC patients compared to healthy controls. We identified 289 DEGs between the DNT populations of both groups. GO and KEGG pathway analyses revealed that the DEGs were mainly related to immunoglobulin mediated immune response, complement activation, and B cell receptor signaling. The PPI networks of the common DEGs were constructed using Cytoscape, and 10 core genes were identified, including TMEM176B, C1QB, C1QC, RASD2, and IFIT3. The expression levels of these genes correlated with the prognosis and immune infiltration in BC patients, and were validated by RT-qPCR (P < 0.05). CONCLUSIONS DNT cells are abundant in patients with BC, and might exert anti-tumor immune responses by regulating genes such as TMEM176B and EGR1.
Collapse
Affiliation(s)
- Hui-Ru Zhu
- School of Medical Laboratory, Shandong Second Medical University, Weifang, China
- Department of Laboratory Medicine, the 960, Hospital of the PLA Joint Logistics Support Force, Jinan, 250031, China
| | - Yun-Bo Wei
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Jia-Qi Guo
- School of Medical Laboratory, Shandong Second Medical University, Weifang, China
| | - Xiao-Fei Liu
- Department of Laboratory Medicine, the 960, Hospital of the PLA Joint Logistics Support Force, Jinan, 250031, China.
| |
Collapse
|
13
|
Beccacece I, Burstein VL, Almeida MA, Gareca JC, Guasconi L, Mena CJ, Mary VS, Theumer MG, Cervi L, Prinz I, Gruppi A, Lionakis MS, Chiapello LS. Cutaneous Innate Lymphoid Populations Drive IL-17A-Mediated Immunity in Nannizzia gypsea Dermatophytosis. J Invest Dermatol 2024:S0022-202X(24)03029-X. [PMID: 39722247 DOI: 10.1016/j.jid.2024.11.011] [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: 04/10/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 12/28/2024]
Abstract
Fungal skin infections significantly contribute to the global human disease burden, yet our understanding of cutaneous immunity against dermatophytes remains limited. Previously, we developed a model of epicutaneous infection with Microsporum canis in C57BL/6 mice, which highlighted the critical role of IL-17RA signaling in antidermatophyte defenses. In this study, we expanded our investigation to the human pathogen Nannizzia gypsea and demonstrated that skin γδTCRint and CD8/CD4 double-negative βTCR+ T cells are the principal producers of IL-17A during dermatophytosis. These IL-17A+ T cells exhibited an activated/memory phenotype, including a subset of proliferating tissue-resident cells. Notably, restriction of lymphocyte trafficking after fingolimod administration in infected mice did not lead to increased susceptibility, indicating that local antifungal defenses are independent of T-cell priming in lymph nodes. In addition, Rag1-/- mice lacking T and B lymphocytes effectively controlled infection and exhibited increased IL-17A production by innate lymphoid cells. Furthermore, Rag2-/-Il2rg-/- mice, devoid of T, B, and innate lymphoid cells, were highly susceptible to dermatophytosis compared with Rag2-/-or wild-type mice, demonstrating that innate lymphoid cells are sufficient to antifungal defenses in T-cell-deficient mice. In conclusion, our study underscores the coordinated interplay between skin γδT, αβT, and innate lymphoid cell subsets in controlling primary N gypsea dermatophytosis.
Collapse
Affiliation(s)
- Ignacio Beccacece
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), National Scientific and Technical Research Council (CONICET), Córdoba, Argentina
| | - Verónica Liliana Burstein
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), National Scientific and Technical Research Council (CONICET), Córdoba, Argentina
| | - Mariel Abigail Almeida
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), National Scientific and Technical Research Council (CONICET), Córdoba, Argentina
| | - Julio Cesar Gareca
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), National Scientific and Technical Research Council (CONICET), Córdoba, Argentina
| | - Lorena Guasconi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), National Scientific and Technical Research Council (CONICET), Córdoba, Argentina
| | - Cristian Javier Mena
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), National Scientific and Technical Research Council (CONICET), Córdoba, Argentina
| | - Verónica Sofia Mary
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), National Scientific and Technical Research Council (CONICET), Córdoba, Argentina
| | - Martín Gustavo Theumer
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), National Scientific and Technical Research Council (CONICET), Córdoba, Argentina
| | - Laura Cervi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), National Scientific and Technical Research Council (CONICET), Córdoba, Argentina
| | - Immo Prinz
- Zentrum für Molekulare Neurobiologie Hamburg, Institut für Systemimmunologie/Institute of Systems Immunology, Medical Center Hamburg-Eppendorf. Falkenried, Germany
| | - Adriana Gruppi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), National Scientific and Technical Research Council (CONICET), Córdoba, Argentina
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Laura Silvina Chiapello
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), National Scientific and Technical Research Council (CONICET), Córdoba, Argentina.
| |
Collapse
|
14
|
Dossybayeva K, Zhubanova G, Mussayeva A, Mukusheva Z, Dildabayeva A, Nauryzbayeva G, Akhmaltdinova L, Orumbayeva U, Tanko M, Poddighe D. Nonspecific increase of αβTCR + double-negative T cells in pediatric rheumatic diseases. World J Pediatr 2024; 20:1283-1292. [PMID: 39604769 PMCID: PMC11634929 DOI: 10.1007/s12519-024-00854-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/17/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND An increased number of double-negative T (DNT) cells expressing the αβ T cell receptor (αβ+DNT cells) is one of the diagnostic criteria for autoimmune lymphoproliferative syndrome (ALPS). Moreover, these cells are expanded in a widely used murine model for lupus. However, the homeostasis of αβ+DNT cells remains inadequately investigated in rheumatic disorders, especially in pediatric patients. METHODS In this cross-sectional, prospective, and observational study, children with rheumatic disorders and healthy controls were recruited to analyze the quantity and characteristics of circulating DNT cells using flow cytometry. RESULTS Overall, the two study groups did not differ in their total DNT cell pool in the bloodstream. However, the number of αβ+DNT cells was significantly higher in rheumatic children than that in the controls, whereas the γδ+DNT cells remained similar. This expansion in the circulating pool of αβ+DNT cells was comparable across different rheumatic diseases, all showing significant differences from the controls in this regard. Moreover, no significant correlation was found between αβ+DNT cell numbers and disease activity. CONCLUSIONS These preliminary results indicate that circulating αβ+DNT cells are significantly expanded in children with rheumatic disorders; however, this finding appears to be a nonspecific (disease-unrelated) marker of autoimmunity. Further and larger studies are necessary to better investigate and define the role of DNT cells in pediatric rheumatic diseases.
Collapse
Affiliation(s)
- Kuanysh Dossybayeva
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), 010000, Astana, Kazakhstan
| | - Gulsamal Zhubanova
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), 010000, Astana, Kazakhstan
| | - Assel Mussayeva
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), 010000, Astana, Kazakhstan
| | - Zaure Mukusheva
- Program of Pediatric Rheumatology, Clinical Academic Department of Pediatrics, University Medical Center, Astana, Kazakhstan
| | - Aiken Dildabayeva
- Program of Pediatric Rheumatology, Clinical Academic Department of Pediatrics, University Medical Center, Astana, Kazakhstan
| | - Galiya Nauryzbayeva
- Clinical Academic Department of Laboratory Medicine, Pathology and Genetics, Republican Diagnostic Center, University Medical Center, Astana, Kazakhstan
| | - Lyudmila Akhmaltdinova
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health (NRCMCH), University Medical Center(UMC), 010000, Astana, Kazakhstan
| | - Ulbolsyn Orumbayeva
- Clinical Academic Department of Laboratory Medicine, Pathology and Genetics, Republican Diagnostic Center, University Medical Center, Astana, Kazakhstan
| | - Matthew Tanko
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), 010000, Astana, Kazakhstan
- Clinical Academic Department of Laboratory Medicine, Pathology and Genetics, Republican Diagnostic Center, University Medical Center, Astana, Kazakhstan
| | - Dimitri Poddighe
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), 010000, Astana, Kazakhstan.
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health (NRCMCH), University Medical Center(UMC), 010000, Astana, Kazakhstan.
- College of Health Sciences, VinUniversity, Gia Lam District, Hanoi, Vietnam.
| |
Collapse
|
15
|
Poddighe D, Maulenkul T, Dossybayeva K, Zhubanova G, Mukusheva Z, Akhmaltdinova L. Double-negative T cells in pediatric rheumatic diseases. Clin Exp Pediatr 2024; 67:632-640. [PMID: 39265625 PMCID: PMC11621738 DOI: 10.3345/cep.2023.01760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 09/14/2024] Open
Abstract
Double-negative (CD4-CD8-) T (DNT) cells have been implicated in autoimmune lymphoproliferative syndrome (ALPS), where their expansion inside the circulating pool of T cells represents a diagnostic criterion. Recent experimental evidence has supported the immunomodulatory roles of DNT cells, and studies in adult patients have suggested that they may be altered in some immune-mediated conditions. This study aimed to retrieve available data on circulating DNT cells in pediatric rheumatic disorders that do not arise in the context of ALPS through a systematic literature review of 3 scientific databases (PubMed, Scopus, and Web of Science). The final output of the systematic literature search consisted of 8 manuscripts, including cross-sectional (n=6) and longitudinal (n=2) studies. Overall, the pooled population of patients includes children affected with pediatric systemic lupus erythematosus (n= 104), juvenile idiopathic arthritis (n=92), Behçet's disease (n=15), mixed connective tissue disease (n=8), juvenile dermatomyositis (n=6), and Kawasaki disease/multisystem inflammatory disease in children (n=1 and n=14, respectively); moreover, one study also included 11 children with a high titer of antinuclear antibody but no diagnosis of rheumatic disease. All studies except one included a control group. The number of DNT cells were increased in most studies of children with rheumatic diseases. Even if such a limited number of studies and their great heterogeneity in several methodological aspects do not allow for reliable conclusions about the relevance of DNT cells in specific rheumatic conditions in children, this cell population deserves further investigation in this pathological setting through well-designed clinical studies.
Collapse
Affiliation(s)
- Dimitri Poddighe
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), Astana, Kazakhstan
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health, University Medical Center, Astana, Kazakhstan
- College of Health Sciences, VinUniversity, Hanoi, Vietnam
| | - Tilektes Maulenkul
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), Astana, Kazakhstan
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health, University Medical Center, Astana, Kazakhstan
| | - Kuanysh Dossybayeva
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), Astana, Kazakhstan
| | - Gulsamal Zhubanova
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), Astana, Kazakhstan
| | - Zaure Mukusheva
- Program of Pediatric Rheumatology, Clinical Academic Department of Pediatrics, University Medical Center, Astana, Kazakhstan
| | - Lyudmila Akhmaltdinova
- Department of Medicine, Nazarbayev University School of Medicine (NUSOM), Astana, Kazakhstan
| |
Collapse
|
16
|
Ai N, Zhang Y, Yang J, Zhang Y, Zhao X, Feng H. Genetically predicted blood metabolites mediate the association between circulating immune cells and severe COVID-19: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40509. [PMID: 39560514 PMCID: PMC11575977 DOI: 10.1097/md.0000000000040509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024] Open
Abstract
Investigating the causal relationship between circulating immune cells, blood metabolites, and severe COVID-19 and revealing the role of blood metabolite-mediated circulating immune cells in disease onset and progression. Genetic variation data of 731 circulating immune cells, 1400 blood metabolites, and severe COVID-19 from genome-wide association study open-access database (https://gwas.mrcieu.ac.uk) were used as instrumental variables for bidirectional and two-step Mendelian randomization analysis. The study identified 11 circulating immune cells with unidirectional causality to severe COVID-19. Two-step Mendelian randomization analysis showed 10 blood metabolites were causally associated with severe COVID-19, and blood Myristate and Citrulline to phosphate ratio mediated the association of circulating effector memory double negative % DN and CD8dim natural killer T cell % T cells, respectively, with severe COVID-19 (Myristate mediated effect ratio was 10.20%, P = .011; Citrulline to phosphate ratio mediated effect ratio was -9.21%, P = .017). This study provides genetic evidence assessing the causal relationship between circulating immune cells, blood metabolites, and severe COVID-19, elucidates the role of blood metabolite-mediated circulating immune cells in severe COVID-19 development, and offers new insights into severe COVID-19 etiology and related preventive and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ning Ai
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Zhang
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Yang
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Zhang
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuejing Zhao
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huifen Feng
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
17
|
Luan Y, Min Q, Zhang R, Wen Z, Meng X, Hu Z, Feng X, Yu M, Dong L, Wang JY. EAF2 deficiency attenuates autoimmune disease in Fas lpr mice by modulating B cell activation and apoptosis. iScience 2024; 27:111220. [PMID: 39555413 PMCID: PMC11565555 DOI: 10.1016/j.isci.2024.111220] [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: 07/15/2024] [Revised: 10/06/2024] [Accepted: 10/17/2024] [Indexed: 11/19/2024] Open
Abstract
MRL/lpr mice develop systemic lupus erythematosus-like autoimmunity due to defective FAS-mediated apoptosis. We generated Fas lpr mice deficient in EAF2, a transcription elongation-associated factor known to promote apoptosis in germinal center (GC) B cells and crucial for preventing autoimmunity. Contrary to expectations, EAF2 deficiency significantly reduced lymphadenopathy and splenomegaly, extended lifespan, and alleviated nephritis by decreasing renal immune complex deposition. Additionally, EAF2 deficiency markedly reduced accumulation of activated B cells, GC B cells, plasma cells, and the abnormal B220+CD3+ T cells in Fas lpr mice. Further analysis revealed that Eaf2 -/- Fas lpr B cells showed hyperactivation upon various stimulations, followed by increased death. RNA sequencing of the B220+CD3+ cells revealed a downregulation in survival-promoting genes such as Bcl-2 and Akt and an upregulation of proapoptotic genes. We conclude that the combined deficiency in FAS- and EAF2-mediated apoptotic pathways leads to B cell hyperactivation and subsequent death, thereby ameliorating systemic autoimmunity in this model.
Collapse
Affiliation(s)
- Yingying Luan
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qing Min
- Department of Clinical Immunology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China
| | - Runyun Zhang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zichao Wen
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xin Meng
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ziying Hu
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiaoqian Feng
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meiping Yu
- Department of Clinical Immunology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
| | - Lulu Dong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Clinical Immunology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China
| |
Collapse
|
18
|
Koh CC, Velikkakam T, Neves EGA, Medeiros NI, Gomes JA, Silva SDA, Gollob KJ, Nunes MDCP, Dutra WO. TNF-expressing CD1d+ monocytes are associated with the activation of CD4- CD8- T cells in patients with Chagas cardiomyopathy. Rev Soc Bras Med Trop 2024; 57:S0037-86822024000100713. [PMID: 39570155 DOI: 10.1590/0037-8682-0181-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/02/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Chagas disease cardiomyopathy is characterized by intense immune activation, with double-negative (DN) T cells as key producers of inflammatory cytokines. CD1d is an antigen-presenting molecule involved in the activation of DN T cells. METHODS We characterized CD1d+ monocytes from patients with cardiac (CARD) and indeterminate (IND) disease using flow cytometry. RESULTS CARD CD1d+ monocytes exhibited higher expression of TNF, TNF-receptor, PDL-1, and Fas-L compared to those from IND. These monocytes correlated with TNF expression by DN T-cells in CARD but not in IND. CONCLUSIONS CD1d+ monocytes from CARD are inflammatory and associated with DN T-cell activation, confirming that CD1d is a target for modulating inflammation in Chagas cardiomyopathy.
Collapse
Affiliation(s)
- Carolina Cattoni Koh
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Belo Horizonte, MG, Brasil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, BA, Brasil
| | - Teresiama Velikkakam
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Belo Horizonte, MG, Brasil
| | | | - Nayara Ingrid Medeiros
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Belo Horizonte, MG, Brasil
| | - Juliana Assis Gomes
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Belo Horizonte, MG, Brasil
| | | | - Kenneth John Gollob
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, BA, Brasil
- Hospital Israelita Albert Einstein, São Paulo, SP, Brasil
| | | | - Walderez Ornelas Dutra
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Belo Horizonte, MG, Brasil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Salvador, BA, Brasil
| |
Collapse
|
19
|
Baez A, Singh D, He S, Hajiaghayi M, Gholizadeh F, Darlington PJ, Helfield B. Immunomodulation of human T cells by microbubble-mediated focused ultrasound. Front Immunol 2024; 15:1486744. [PMID: 39502696 PMCID: PMC11534865 DOI: 10.3389/fimmu.2024.1486744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
While met with initial and ground-breaking success targeting blood borne cancers, cellular immunotherapy remains significantly hindered in the context of solid tumors by the tumor microenvironment. Focused ultrasound, in conjunction with microbubbles, has found tremendous potential as a targeted and local drug/gene delivery technique for cancer therapy. The specific immunomodulating effects of this technique on immune cells, including T-cells, remain unexplored. Here, with freshly isolated human immune cells, we examine how focused ultrasound can viably modulate immune cell membrane permeability and influence the secretion of over 90 cytokines, chemokines and other analytes relevant to a potent immune response against cancer. We determine that microbubble-mediated focused ultrasound modulates the immune cell secretome in a time-dependent manner - ranging in ~0.1-3.6-fold changes in the concentration of a given cytokine compared to sham controls over 48 hours post-treatment (e.g. IL-1β, TNF-α, CX3CL1, CCL21). Further, we determine the general trend of a negative correlation between secreted cytokine concentration and viable ultrasound-assisted membrane permeability with negligible loss of cell viability. Taken together, the data presented here highlights the potential of microbubble-mediated focused ultrasound to viably enhance T-cell permeability and modulate key pro-immune pathways, offering a novel approach to augment targeted cellular therapies for solid tumors.
Collapse
Affiliation(s)
- Ana Baez
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Davindra Singh
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Stephanie He
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Mehri Hajiaghayi
- Department of Biology, Concordia University, Montreal, QC, Canada
| | | | | | - Brandon Helfield
- Department of Biology, Concordia University, Montreal, QC, Canada
- Department of Physics, Concordia University, Montreal, QC, Canada
| |
Collapse
|
20
|
Yero A, Shi T, Clain JA, Zghidi-Abouzid O, Racine G, Costiniuk CT, Routy JP, Estaquier J, Jenabian MA. Double-Negative T-Cells during Acute Human Immunodeficiency Virus and Simian Immunodeficiency Virus Infections and Following Early Antiretroviral Therapy Initiation. Viruses 2024; 16:1609. [PMID: 39459942 PMCID: PMC11512404 DOI: 10.3390/v16101609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
HIV infection significantly affects the frequencies and functions of immunoregulatory CD3+CD4-CD8- double-negative (DN) T-cells, while the effect of early antiretroviral therapy (ART) initiation on these cells remains understudied. DN T-cell subsets were analyzed prospectively in 10 HIV+ individuals during acute infection and following early ART initiation compared to 20 HIV-uninfected controls. In this study, 21 Rhesus macaques (RMs) were SIV-infected, of which 13 were assessed during acute infection and 8 following ART initiation four days post-infection. DN T-cells and FoxP3+ DN Treg frequencies increased during acute HIV infection, which was not restored by ART. The expression of activation (HLA-DR/CD38), immune checkpoints (PD-1/CTLA-4), and senescence (CD28-CD57+) markers by DN T-cells and DN Tregs increased during acute infection and was not normalized by ART. In SIV-infected RMs, DN T-cells remained unchanged despite infection or ART, whereas DN Treg frequencies increased during acute SIV infection and were not restored by ART. Finally, frequencies of CD39+ DN Tregs increased during acute HIV and SIV infections and remained elevated despite ART. Altogether, acute HIV/SIV infections significantly changed DN T-cell and DN Treg frequencies and altered their immune phenotype, while these changes were not fully normalized by early ART, suggesting persistent HIV/SIV-induced immune dysregulation despite early ART initiation.
Collapse
Affiliation(s)
- Alexis Yero
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC H2X 3X8, Canada; (A.Y.); (T.S.)
| | - Tao Shi
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC H2X 3X8, Canada; (A.Y.); (T.S.)
| | - Julien A. Clain
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (O.Z.-A.); (G.R.); (J.E.)
| | - Ouafa Zghidi-Abouzid
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (O.Z.-A.); (G.R.); (J.E.)
| | - Gina Racine
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (O.Z.-A.); (G.R.); (J.E.)
| | - Cecilia T. Costiniuk
- Infectious Diseases and Immunity in Global Health Program, Research Institute of McGill University Health Centre, Montreal, QC H3H 2R9, Canada; (C.T.C.); (J.-P.R.)
- Chronic Viral Illness Service, Division of Infectious Disease, Department of Medicine, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Jean-Pierre Routy
- Infectious Diseases and Immunity in Global Health Program, Research Institute of McGill University Health Centre, Montreal, QC H3H 2R9, Canada; (C.T.C.); (J.-P.R.)
- Chronic Viral Illness Service, Division of Infectious Disease, Department of Medicine, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Jérôme Estaquier
- Centre Hospitalier Universitaire (CHU) de Québec Centre de Recherche, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; (J.A.C.); (O.Z.-A.); (G.R.); (J.E.)
| | - Mohammad-Ali Jenabian
- Department of Biological Sciences and CERMO-FC Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC H2X 3X8, Canada; (A.Y.); (T.S.)
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
21
|
Yang W, Ou Y, Luo H, You L, Du H. Causal relationship between circulating immune cells and gastric cancer: a bidirectional Mendelian randomization analysis using UK Biobank and FinnGen datasets. Transl Cancer Res 2024; 13:4702-4713. [PMID: 39430856 PMCID: PMC11483344 DOI: 10.21037/tcr-24-480] [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: 03/24/2024] [Accepted: 08/01/2024] [Indexed: 10/22/2024]
Abstract
Background The role of immune cells in cancer pathogenesis remains controversial due to conflicting reports, potentially arising from various confounding factors. Emerging evidence suggests that cancer can also influence immune cell populations and functions, making it challenging to investigate their causal relationship. Traditional observational studies often fail to eliminate all confounding factors and are prone to reverse causality. Therefore, we employ Mendelian randomization (MR) to determine the causal relationship between immune cells and cancer, as this method can identify causal relationships independent of confounding factors and avoid reverse causality. Methods Genome-wide association study (GWAS) summary statistics on immune traits, encompassing 310 immune cell phenotypes, were obtained from 3,757 European individuals, with peripheral blood immune cells tested using flow cytometry. GWAS summary statistics for gastric cancer were derived from 476,116 European individuals across two large-scale biobanks: the UK Biobank and FinnGen. Gastric cancer was identified by the International Classification of Diseases, 9th Revision (ICD-9), and 10th Revision (ICD-10) codes. Significant single nucleotide polymorphisms (SNPs) for immune traits were extracted at a threshold of P<1×10-5, while a threshold of P<5×10-8 was used for gastric cancer GWAS data. Linkage imbalance-based clumping was performed to obtain independent SNPs, and those with F<10 were excluded to mitigate weak instrument bias. Phenoscanner V2 was used to exclude SNPs directly associated with potential confounders or outcomes. Two-sample MR was conducted using five MR methods, with inverse-variance-weighted (IVW) as the primary analysis method. A false discovery rate (FDR) correction was used to reduce the likelihood of type 1 errors. In addition, we conducted MR-Egger intercept tests and Cochran's Q tests. Results The numbers of CD4-CD8- T cells and IgD-CD27- B cells were positively correlated with the development of gastric cancer, with odds ratios (ORs) of 1.15 [95% confidence interval (CI), 1.07-1.24; P<0.001; PFDR=0.041; IVW method] and 1.07 (95% CI, 1.03-1.11; P=0.001; PFDR=0.187; IVW method), respectively. However, the percentage of IgD+CD24- B cells in lymphocytes were negatively associated with the development of gastric cancer (OR =0.90; 95% CI, 0.84-0.96; P=0.002; PFDR=0.187; IVW method). MR analysis of the above three immune cell phenotypes showed no significant heterogeneity or horizontal pleiotropy. In the reverse MR analysis, gastric cancer was not causally associated with any of the immune cell phenotypes. Conclusions Circulating CD4-CD8- T cells and IgD-CD27- B cells are positively correlated with the development of gastric cancer, while the percentage of IgD+CD24- B cells in lymphocytes are negatively correlated. These findings provide insight into the relationship between immune cells and gastric cancer pathogenesis and may serve as a basis for the development of immunotherapies for gastric cancer.
Collapse
Affiliation(s)
- Weimin Yang
- Department of Surgery, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, China
| | - Yang Ou
- Department of Surgery, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, China
| | - Hui Luo
- Department of Gastrointestinal Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Lijuan You
- Department of Anesthesiology, Huanggang Central Hospital Affiliated to Yangtze University, Huanggang, China
| | - Heng Du
- Department of Gastrointestinal Surgery, Huanggang Central Hospital Affiliated to Yangtze University, Huanggang, China
| |
Collapse
|
22
|
Cimerman M, Druzhaeva N, Nemec Svete A, Hajdinjak M, Pohar K, Ihan A, Domanjko Petrič A. Inflammatory and immune variables as predictors of survival in dogs with myxomatous mitral valve disease. BMC Vet Res 2024; 20:431. [PMID: 39342325 PMCID: PMC11438168 DOI: 10.1186/s12917-024-04266-7] [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: 06/07/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND We aimed to investigate the association between selected inflammatory and immune variables and survival of dogs with myxomatous mitral valve disease (MMVD). We evaluated data of 62 client-owned dogs with MMVD, grouped into preclinical, stable congestive heart failure (CHF) and unstable CHF. Univariate Cox proportional hazards regression analysis was used to quantify the association of white blood cell count, concentrations and percentages of T lymphocytes and their subtypes (T helper lymphocytes, cytotoxic T lymphocytes, double positive T lymphocytes, double negative T lymphocytes) and B lymphocytes with survival. P values < 0.1 in individual groups and P values < 0.05 in the group of all patients were considered significant. Spearman correlation coefficients between significant covariates were calculated to assess the relationships among variables and with survival. RESULTS In the preclinical group, percentage of double positive T lymphocytes was negatively associated with survival (hazard ratio (HR) = 2.328; P = 0.051). In the unstable CHF, T lymphocyte (HR = 1.613; P = 0.085), cytotoxic T lymphocyte (HR = 1.562; P = 0.048), double positive (HR = 1.751; P = 0.042), and double negative T lymphocyte (HR = 1.613; P = 0.096) concentrations were negatively associated with survival, as well as cytotoxic T lymphocyte (HR = 1.502; P = 0.007) concentration in the group of all patients. The percentage of T helper lymphocytes was positively associated with survival in the unstable CHF (HR = 0.604; P = 0.053) and in the group of all patients (HR = 0.733; P = 0.044). The concentration of cytotoxic T lymphocytes positively correlated with left atrial to aortic ratio (LA/Ao) (rho = 0.259, P = 0.037), and peak velocity of early diastolic mitral flow (rho = 0.259, P = 0.039), whereas the percentage of T helper lymphocytes negatively correlated with left atrial to aortic ratio (LA/Ao) (rho = -0.212, P = 0.090) and early to late mitral flow ratio (rho = -0.232, P = 0.072). CONCLUSIONS Cytotoxic T lymphocytes, T helper lymphocytes, double positive and double negative T lymphocytes as well as biomarkers cardiac troponin I, N-terminal pro-B-type natriuretic peptide, C-reactive protein are implicated in the progression of MMVD.
Collapse
Affiliation(s)
- Martina Cimerman
- Veterinary Faculty, Small Animal Clinic, University of Ljubljana, Ljubljana, Slovenia
| | - Natalia Druzhaeva
- Veterinary Faculty, Small Animal Clinic, University of Ljubljana, Ljubljana, Slovenia
| | - Alenka Nemec Svete
- Veterinary Faculty, Small Animal Clinic, University of Ljubljana, Ljubljana, Slovenia
| | - Melita Hajdinjak
- Faculty of Electrical Engineering, Laboratory of Applied Mathematics and Statistics, University of Ljubljana, Ljubljana, Slovenia
| | - Katka Pohar
- Faculty of Medicine, Institute of Microbiology and Immunology, University of Ljubljana, Ljubljana, Slovenia
| | - Alojz Ihan
- Faculty of Medicine, Institute of Microbiology and Immunology, University of Ljubljana, Ljubljana, Slovenia
| | | |
Collapse
|
23
|
Macchia I, La Sorsa V, Ciervo A, Ruspantini I, Negri D, Borghi M, De Angelis ML, Luciani F, Martina A, Taglieri S, Durastanti V, Altavista MC, Urbani F, Mancini F. T Cell Peptide Prediction, Immune Response, and Host-Pathogen Relationship in Vaccinated and Recovered from Mild COVID-19 Subjects. Biomolecules 2024; 14:1217. [PMID: 39456150 PMCID: PMC11505848 DOI: 10.3390/biom14101217] [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: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
COVID-19 remains a significant threat, particularly to vulnerable populations. The emergence of new variants necessitates the development of treatments and vaccines that induce both humoral and cellular immunity. This study aimed to identify potentially immunogenic SARS-CoV-2 peptides and to explore the intricate host-pathogen interactions involving peripheral immune responses, memory profiles, and various demographic, clinical, and lifestyle factors. Using in silico and experimental methods, we identified several CD8-restricted SARS-CoV-2 peptides that are either poorly studied or have previously unreported immunogenicity: fifteen from the Spike and three each from non-structural proteins Nsp1-2-3-16. A Spike peptide, LA-9, demonstrated a 57% response rate in ELISpot assays using PBMCs from 14 HLA-A*02:01 positive, vaccinated, and mild-COVID-19 recovered subjects, indicating its potential for diagnostics, research, and multi-epitope vaccine platforms. We also found that younger individuals, with fewer vaccine doses and longer intervals since infection, showed lower anti-Spike (ELISA) and anti-Wuhan neutralizing antibodies (pseudovirus assay), higher naïve T cells, and lower central memory, effector memory, and CD4hiCD8low T cells (flow cytometry) compared to older subjects. In our cohort, a higher prevalence of Vδ2-γδ and DN T cells, and fewer naïve CD8 T cells, seemed to correlate with strong cellular and lower anti-NP antibody responses and to associate with Omicron infection, absence of confusional state, and habitual sporting activity.
Collapse
Affiliation(s)
- Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina La Sorsa
- Research Promotion and Coordination Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Alessandra Ciervo
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Irene Ruspantini
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Francesca Luciani
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Antonio Martina
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Silvia Taglieri
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina Durastanti
- Neurology Unit, San Filippo Neri Hospital, ASL RM1, 00135 Rome, Italy; (V.D.); (M.C.A.)
| | | | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Fabiola Mancini
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| |
Collapse
|
24
|
Tabata R, Tabata C. Possible Association of CD3+CD4-CD8- Phenotype of T-Cell Lymphoma with Peripheral Blood Eosinophilia. Int Arch Allergy Immunol 2024; 186:184-188. [PMID: 39278207 DOI: 10.1159/000541097] [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: 07/15/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024] Open
Abstract
INTRODUCTION Because the prognosis of patients with peripheral T-cell lymphoma is poor compared to that of patients with B-cell lymphoma, we want to avoid further organ damage by eosinophilia. Moreover, in patients with some types of lymphoma, blood eosinophilia is implicated in a worse prognosis. To study the risk factors of eosinophilia, the association between lymphoma type, immunophenotypic features, and peripheral blood eosinophil counts were examined in the patients with mature T-cell lymphoma. METHODS We retrospectively examined 28 patients with mature T-cell lymphoma who were admitted to our hospital and whose immunophenotypic features were confirmed using flow cytometric, immunohistochemical analysis, or both between December 2012 and November 2023. RESULTS We report a possible association between peripheral eosinophilia and peripheral T-cell lymphoma - not otherwise specified and CD3+CD4-D8- (double-negative) phenotypes. Mild eosinophilia was observed in various types, but moderate and severe eosinophilia were observed in patients with peripheral T-cell lymphoma - not otherwise specified. Double-negative phenotype was rarely observed; however, all patients with double-negative phenotype exhibited peripheral blood eosinophilia. In addition, four of the five cases of the double-negative type were peripheral T-cell lymphoma - not otherwise specified. CONCLUSION Here, we retrospectively examined patients with peripheral T-cell lymphoma whose immunophenotypic features were confirmed and report a possible association between peripheral eosinophilia and peripheral T-cell lymphoma - not otherwise specified and CD3+CD4-CD8- (double-negative) phenotypes. In addition, clinicians should be aware of the possible risk that patients with lymphocytic hypereosinophilic syndrome of the double-negative phenotype may develop peripheral T-cell lymphoma.
Collapse
Affiliation(s)
- Rie Tabata
- Department of Hematology, Saiseikai Noe Hospital, Osaka, Japan
| | - Chiharu Tabata
- Department of Pharmacy, School of Pharmacy, Hyogo Medical University, Hyogo, Japan
| |
Collapse
|
25
|
de Matos Rodrigues J, Lokhande L, Olsson LM, Hassan M, Johansson A, Janská A, Kumar D, Schmidt L, Nikkarinen A, Hollander P, Glimelius I, Porwit A, Gerdtsson AS, Jerkeman M, Ek S. CD163+ macrophages in mantle cell lymphoma induce activation of prosurvival pathways and immune suppression. Blood Adv 2024; 8:4370-4385. [PMID: 38959399 PMCID: PMC11375268 DOI: 10.1182/bloodadvances.2023012039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024] Open
Abstract
ABSTRACT Mantle cell lymphoma (MCL) is dependent on a supportive tumor immune microenvironment (TIME) in which infiltration of CD163+ macrophages has a negative prognostic impact. This study explores how abundance and spatial localization of CD163+ cells are associated with the biology of MCL, using spatial multiomic investigations of tumor and infiltrating CD163+ and CD3+ cells. A total of 63 proteins were measured using GeoMx digital spatial profiling in tissue microarrays from 100 diagnostic MCL tissues. Regions of interest were selected in tumor-rich and tumor-sparse tissue regions. Molecular profiling of CD163+ macrophages, CD20+ MCL cells, and CD3+ T-cells was performed. To validate protein profiles, 1811 messenger RNAs were measured in CD20+ cells and 2 subsets of T cells. Image analysis was used to extract the phenotype and position of each targeted cell, thereby allowing the exploration of cell frequencies and cellular neighborhoods. Proteomic investigations revealed that CD163+ cells modulate their immune profile depending on their localization and that the immune inhibitory molecules, V-domain immunoglobulin suppressor of T-cell activation and B7 homolog 3, have higher expression in tumor-sparse than in tumor-rich tissue regions and that targeting should be explored. We showed that MCL tissues with more abundant infiltration of CD163+ cells have a higher proteomic and transcriptional expression of key components of the MAPK pathway. Thus, the MAPK pathway may be a feasible therapeutic target in patients with MCL with CD163+ cell infiltration. We further showed the independent and combined prognostic values of CD11c and CD163 beyond established risk factors.
Collapse
Affiliation(s)
| | | | - Lina M. Olsson
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - May Hassan
- Department of Immunotechnology, Lund University, Lund, Sweden
| | | | - Anna Janská
- Department of Immunotechnology, Lund University, Lund, Sweden
| | | | - Lina Schmidt
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Anna Nikkarinen
- Department of Immunology, Genetics and Pathology, Cancer Precision Medicine, Uppsala University, Uppsala, Sweden
| | - Peter Hollander
- Department of Immunology, Genetics and Pathology, Clinical and Experimental Pathology, Uppsala University, Uppsala, Sweden
| | - Ingrid Glimelius
- Department of Immunology, Genetics and Pathology, Cancer Precision Medicine, Uppsala University, Uppsala, Sweden
| | - Anna Porwit
- Division of Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | | | - Mats Jerkeman
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Sara Ek
- Department of Immunotechnology, Lund University, Lund, Sweden
| |
Collapse
|
26
|
Karwig L, Moore PF, Alber G, Eschke M. Distinct characteristics of unique immunoregulatory canine non-conventional TCRαβ pos CD4 negCD8α neg double-negative T cell subpopulations. Front Immunol 2024; 15:1439213. [PMID: 39185407 PMCID: PMC11341405 DOI: 10.3389/fimmu.2024.1439213] [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: 05/27/2024] [Accepted: 07/12/2024] [Indexed: 08/27/2024] Open
Abstract
Conventional CD4pos regulatory T (Treg) cells characterized by expression of the key transcription factor forkhead box P3 (FoxP3) are crucial to control immune responses, thereby maintaining homeostasis and self-tolerance. Within the substantial population of non-conventional T cell receptor (TCR)αβpos CD4negCD8αneg double-negative (dn) T cells of dogs, a novel FoxP3pos Treg-like subset was described that, similar to conventional CD4pos Treg cells, is characterized by high expression of CD25. Noteworthy, human and murine TCRαβpos regulatory dn T cells lack FoxP3. Immunosuppressive capacity of canine dn T cells was hypothesized based on expression of inhibitory molecules (interleukin (IL)-10, cytotoxic T-lymphocyte associated protein 4, CTLA4). Here, to verify their regulatory function, the dnCD25pos (enriched for FoxP3pos Treg-like cells) and the dnCD25neg fraction, were isolated by fluorescence-activated cell sorting from peripheral blood mononuclear cells (PBMC) of Beagle dogs and analyzed in an in vitro suppression assay in comparison to conventional CD4posCD25pos Treg cells (positive control) and CD4posCD25neg T cells (negative control). Canine dnCD25pos T cells suppressed the Concanavalin A-driven proliferation of responder PBMC to a similar extent as conventional CD4posCD25pos Treg cells. Albeit to a lesser extent than FoxP3-enriched dn and CD4posCD25pos populations, even dnCD25neg T cells reduced the proliferation of responder cells. This is remarkable, as dnCD25neg T cells have a FoxP3neg phenotype comparable to non-suppressive CD4posCD25neg T cells. Both, CD25pos and CD25neg dn T cells, can mediate suppression independent of cell-cell contact and do not require additional signals from CD4posCD25neg T cells to secrete inhibitory factors in contrast to CD4posCD25pos T cells. Neutralization of IL-10 completely abrogated the suppression by dnCD25pos and CD4posCD25pos Treg cells in a Transwell™ system, while it only partially reduced suppression by dnCD25neg T cells. Taken together, unique canine non-conventional dnCD25pos FoxP3pos Treg-like cells are potent suppressor cells in vitro. Moreover, inhibition of proliferation of responder T cells by the dnCD25neg fraction indicates suppressive function of a subset of dn T cells even in the absence of FoxP3. The identification of unique immunoregulatory non-conventional dn T cell subpopulations of the dog in vitro is of high relevance, given the immunotherapeutic potential of manipulating regulatory T cell responses in vivo.
Collapse
Affiliation(s)
- Laura Karwig
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Peter F. Moore
- Department of Veterinary Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Gottfried Alber
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Maria Eschke
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| |
Collapse
|
27
|
Liu XC, Sun KN, Zhu HR, Dai YL, Liu XF. Diagnostic and prognostic value of double-negative T cells in colorectal cancer. Heliyon 2024; 10:e34645. [PMID: 39114054 PMCID: PMC11305275 DOI: 10.1016/j.heliyon.2024.e34645] [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: 09/04/2023] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
Objective To evaluate the T-lymphocyte subset distribution and the diagnostic and prognosis value of double-negative T (DNT) cells in colorectal cancer (CRC). Methods This retrospective study compared the T-lymphocyte subsets and DNT of 114 patients with CRC with those of 107 healthy controls (HC). The diagnostic potential of DNT and T-lymphocyte subsets was assessed using the receiver operating characteristic (ROC) curve, and prognostic values were evaluated using the Kaplan-Meier curve and the Cox regression model. Results The percentages of CD8+ T cells and DNT cells, and value of carcinoembryonic antigen (CEA), were remarkably higher in patients with CRC than in those with HC, but the ratio of CD4+/CD8+ was decreased. Using ROC curve analysis, DNT cell percentage, CEA, and CD4+/CD8+ ratio all had good diagnostic efficacy, with areas under the curve (AUCs) of 0.865, 0.786 and 0.624, respectively. The combination of DNT cell percentage and CEA had an AUC of 0.905, which was significantly higher than that of any single biomarker (p < 0.05). In univariate analysis, the Tumor Node Metastasis (TNM) clinical stage, CD4+/CD8+ ratio, and DNT cell percentage were significantly associated with overall survival (OS) (p < 0.05). In multivariate analysis, TNM clinical staging (HR = 2.37, 95 % CI: 1.15-4.90), a decreased CD4+/CD8+ ratio (HR = 0.33, 95 % CI: 0.15-0.74), and an increased DNT cell percentage (HR = 2.29, 95 % CI: 1.11-4.73) were independent prognostic factors for CRC. Conclusion The percentage of DNT cells may be useful as an evaluation index for CRC diagnosis and prognosis, which was even better when combined with serum CEA.
Collapse
Affiliation(s)
- Xiao-Cui Liu
- Senior Department of Ophthalmology, the Third Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Ke-Na Sun
- Department of Laboratory Medicine, Weifang People's Hospital, Weifang, 261041, Shandong Province, China
| | - Hui-Ru Zhu
- Department of Laboratory Medicine, Weifang Medical University, Weifang, 261053, Shandong Province, China
| | - Yu-Ling Dai
- Department of Laboratory Medicine, Weifang Medical University, Weifang, 261053, Shandong Province, China
| | - Xiao-Fei Liu
- Department of Laboratory Medicine, the 960th Hospital of the PLA Joint Logistics Support Force, Ji'nan, 250031, Shandong Province, China
| |
Collapse
|
28
|
Chen R, Tao Q, Wu F, Zhai Z, Jiang Y, Xu C, Wang H. DNT cells mediate resistance to CAR-T cells therapy in a pediatric patient with relapsed and refractory B-ALL. Ann Hematol 2024; 103:2551-2556. [PMID: 38724656 DOI: 10.1007/s00277-024-05790-0] [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: 01/18/2024] [Accepted: 05/04/2024] [Indexed: 07/06/2024]
Abstract
Chimeric antigen receptor T (CAR-T) cells therapy is a milestone achievement in the immunotherapy of relapsed and refractory (R/R) B cell acute lymphoblastic leukemia (B-ALL). However, some patients treated with CAR-T cells do not achieve complete remission, the mechanisms of which have not been elucidated. In the present study, we report a 9-year-old pediatric patient with refractory B-ALL received a triple infusion of autologous CD19 CAR-T cells therapy after the second relapse. CAR-T cells expanded in the peripheral blood and bone marrow. However, the patient did not achieve complete remission, indicating a lack of response to CAR-T cells therapy. Analysis of etiological factors revealed that the number of CD4 and CD8 double-negative T (DNT) cells was significantly upregulated in the peripheral blood, bone marrow, and autologous CAR-T cells products. In conclusiont, these findings indicate that DNT cells mediated resistance to CAR-T cells therapy in this pediatric patient with R/R B-ALL.
Collapse
Affiliation(s)
- Ruotong Chen
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qianshan Tao
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fan Wu
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhimin Zhai
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuchen Jiang
- Department of Biotechnology, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | | | - Huiping Wang
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
29
|
Lokhande L, Nilsson D, de Matos Rodrigues J, Hassan M, Olsson LM, Pyl PT, Vasquez L, Porwit A, Gerdtsson AS, Jerkeman M, Ek S. Quantification and Profiling of Early and Late Differentiation Stage T Cells in Mantle Cell Lymphoma Reveals Immunotherapeutic Targets in Subsets of Patients. Cancers (Basel) 2024; 16:2289. [PMID: 39001353 PMCID: PMC11240320 DOI: 10.3390/cancers16132289] [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: 05/02/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/16/2024] Open
Abstract
With the aim to advance the understanding of immune regulation in MCL and to identify targetable T-cell subsets, we set out to combine image analysis and spatial omic technology focused on both early and late differentiation stages of T cells. MCL patient tissue (n = 102) was explored using image analysis and GeoMx spatial omics profiling of 69 proteins and 1812 mRNAs. Tumor cells, T helper (TH) cells and cytotoxic (TC) cells of early (CD57-) and late (CD57+) differentiation stage were analyzed. An image analysis workflow was developed based on fine-tuned Cellpose models for cell segmentation and classification. TC and CD57+ subsets of T cells were enriched in tumor-rich compared to tumor-sparse regions. Tumor-sparse regions had a higher expression of several key immune suppressive proteins, tentatively controlling T-cell expansion in regions close to the tumor. We revealed that T cells in late differentiation stages (CD57+) are enriched among MCL infiltrating T cells and are predictive of an increased expression of immune suppressive markers. CD47, IDO1 and CTLA-4 were identified as potential targets for patients with T-cell-rich MCL TIME, while GITR might be a feasible target for MCL patients with sparse T-cell infiltration. In subgroups of patients with a high degree of CD57+ TC-cell infiltration, several immune checkpoint inhibitors, including TIGIT, PD-L1 and LAG3 were increased, emphasizing the immune-suppressive features of this highly differentiated T-cell subset not previously described in MCL.
Collapse
Affiliation(s)
- Lavanya Lokhande
- Department of Immunotechnology, Lund University, 221 00 Lund, Sweden
| | - Daniel Nilsson
- Department of Immunotechnology, Lund University, 221 00 Lund, Sweden
| | | | - May Hassan
- Department of Immunotechnology, Lund University, 221 00 Lund, Sweden
| | - Lina M. Olsson
- Department of Immunotechnology, Lund University, 221 00 Lund, Sweden
| | - Paul-Theodor Pyl
- Department of Laboratory Medicine, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Lund University, 221 00 Lund, Sweden
| | - Louella Vasquez
- Department of Laboratory Medicine, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Lund University, 221 00 Lund, Sweden
| | - Anna Porwit
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, 221 00 Lund, Sweden
| | | | - Mats Jerkeman
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, 221 00 Lund, Sweden
| | - Sara Ek
- Department of Immunotechnology, Lund University, 221 00 Lund, Sweden
| |
Collapse
|
30
|
Rodríguez-Rodríguez N, Rosetti F, Crispín JC. CD8 is down(regulated) for tolerance. Trends Immunol 2024; 45:442-453. [PMID: 38782625 DOI: 10.1016/j.it.2024.04.012] [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: 03/22/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024]
Abstract
Activated CD8+ T cells directly kill target cells. Therefore, the regulation of their function is central to avoiding immunopathology. Mechanisms that curb effector functions in CD4+ and CD8+ T cells are mostly shared, yet important differences occur. Here, we focus on the control of CD8+ T cell activity and discuss the importance of a poorly understood aspect of tolerance that directly impairs engagement of target cells: the downregulation of CD8. We contextualize this process and propose that it represents a key element during CD8+ T cell modulation.
Collapse
Affiliation(s)
| | - Florencia Rosetti
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José C Crispín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, Mexico.
| |
Collapse
|
31
|
Wen Z, Wang L, Ma H, Li L, Wan L, Shi L, Li H, Chen H, Hao W, Song S, Xue Q, Wei Y, Li F, Xu J, Zhang S, Wong KW, Song Y. Integrated single-cell transcriptome and T cell receptor profiling reveals defects of T cell exhaustion in pulmonary tuberculosis. J Infect 2024; 88:106158. [PMID: 38642678 DOI: 10.1016/j.jinf.2024.106158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/07/2024] [Accepted: 04/12/2024] [Indexed: 04/22/2024]
Abstract
Tuberculosis-affected lungs with chronic inflammation harbor abundant immunosuppressive immune cells but the nature of such inflammation is unclear. Dysfunction in T cell exhaustion, while implicated in chronic inflammatory diseases, remains unexplored in tuberculosis. Given that immunotherapy targeting exhaustion checkpoints exacerbates tuberculosis, we speculate that T cell exhaustion is dysfunctional in tuberculosis. Using integrated single-cell RNA sequencing and T cell receptor profiling we reported defects in exhaustion responses within inflamed tuberculosis-affected lungs. Tuberculosis lungs demonstrated significantly reduced levels of exhausted CD8+ T cells and exhibited diminished expression of exhaustion-related transcripts among clonally expanded CD4+ and CD8+ T cells. Additionally, clonal expansion of CD4+ and CD8+ T cells bearing T cell receptors specific for CMV was observed. Expanded CD8+ T cells expressed the cytolytic marker GZMK. Hence, inflamed tuberculosis-affected lungs displayed dysfunction in T cell exhaustion. Our findings likely hold implications for understanding the reactivation of tuberculosis observed in patients undergoing immunotherapy targeting the exhaustion checkpoint.
Collapse
Affiliation(s)
- Zilu Wen
- Department of Scientific Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Lin Wang
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Hui Ma
- Department of Scientific Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Leilei Li
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Laiyi Wan
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Lei Shi
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Hongwei Li
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Hui Chen
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Wentao Hao
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Shu Song
- Department of Pathology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Qinghua Xue
- Department of Scientific Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yutong Wei
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Feng Li
- Department of Respiratory Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jianqing Xu
- Department of Scientific Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Shulin Zhang
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Ka-Wing Wong
- Department of Scientific Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
| | - Yanzheng Song
- Department of Thoracic Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
| |
Collapse
|
32
|
Xu J, Wang BY, Yu SH, Chen SJ, Yang SS, Liu R, Chen LJ, Hou J, Chen Z, Zhao WH, He AL, Mi JQ, Chen SJ. Long-term remission and survival in patients with relapsed or refractory multiple myeloma after treatment with LCAR-B38M CAR T cells: 5-year follow-up of the LEGEND-2 trial. J Hematol Oncol 2024; 17:23. [PMID: 38659046 PMCID: PMC11040812 DOI: 10.1186/s13045-024-01530-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/20/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND The autologous anti-B-cell maturation antigen (BCMA) chimeric antigen receptor (CAR) T-cell therapy LCAR-B38M has been approved for the treatment of relapsed and refractory multiple myeloma in many countries across the world under the name ciltacabtagene autoleucel. LEGEND-2 was the first-in-human trial of LCAR-B38M and yielded deep and durable therapeutic responses. Here, we reported the outcomes in LEGEND-2 after a minimal 5-year follow-up. METHODS Participants received an average dose of 0.5 × 106 cells/kg LCAR-B38M in split or single unfractionated infusions after cyclophosphamide-based lymphodepletion therapy. Investigator-assessed response, survival, safety and pharmacokinetics were evaluated. RESULTS Seventy-four participants enrolled and had a median follow-up of 65.4 months. The 5-year progression-free survival (PFS) and overall survival (OS) rates were 21.0% and 49.1%, with progressive flattening of the survival curves over time. Patients with complete response (CR) had longer PFS and OS, with 5-year rates of 28.4% and 65.7%, respectively. Twelve patients (16.2%) remained relapse-free irrespective of baseline high-risk cytogenetic abnormality and all had normal humoral immunity reconstituted. An ongoing CR closely correlated with several prognostic baseline indices including favorable performance status, immunoglobulin G subtype, and absence of extramedullary disease, as well as a combination cyclophosphamide and fludarabine preconditioning strategy. Sixty-two (83.8%) suffered progressive disease (PD) and/or death; however, 61.1% of PD patients could well respond to subsequent therapies, among which, the proteasome inhibitor-based regimens benefited the most. Concerning the safety, hematologic and hepatic function recovery were not significantly different between non-PD and PD/Death groups. A low rate of second primary malignancy (5.4%) and no severe virus infection were observed. The patients who tested positive for COVID-19 merely presented self-limiting symptoms. In addition, a sustainable CAR T population of one case with persistent remission was delineated, which was enriched with indolently proliferative and lowly cytotoxic CD4/CD8 double-negative functional T lymphocytes. CONCLUSIONS These data, representing the longest follow-up of BCMA-redirected CAR T-cell therapy to date, demonstrate long-term remission and survival with LCAR-B38M for advanced myeloma. TRIAL REGISTRATION LEGEND-2 was registered under the trial numbers NCT03090659, ChiCTRONH-17012285.
Collapse
Affiliation(s)
- Jie Xu
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Shanghai Institute of Hematology, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Bai-Yan Wang
- Department of Hematology, Second Affiliated Hospital of Xi'an Jiao Tong University, 157 West 5th Road, Xi'an, 710004, China
| | - Shan-He Yu
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Shanghai Institute of Hematology, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Shi-Jun Chen
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Shanghai Institute of Hematology, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Shuang-Shuang Yang
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Shanghai Institute of Hematology, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Rui Liu
- Department of Hematology, Second Affiliated Hospital of Xi'an Jiao Tong University, 157 West 5th Road, Xi'an, 710004, China
| | - Li-Juan Chen
- Department of Hematology, Jiangsu Province Hospital, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jian Hou
- Department of Hematology, Renji Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Zhu Chen
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Shanghai Institute of Hematology, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Wan-Hong Zhao
- Department of Hematology, Second Affiliated Hospital of Xi'an Jiao Tong University, 157 West 5th Road, Xi'an, 710004, China.
| | - Ai-Li He
- Department of Hematology, Second Affiliated Hospital of Xi'an Jiao Tong University, 157 West 5th Road, Xi'an, 710004, China.
| | - Jian-Qing Mi
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Shanghai Institute of Hematology, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China.
| | - Sai-Juan Chen
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Shanghai Institute of Hematology, Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China.
| |
Collapse
|
33
|
Mitchell RA, Ubillos I, Requena P, Campo JJ, Ome-Kaius M, Hanieh S, Umbers A, Samol P, Barrios D, Jiménez A, Bardají A, Mueller I, Menéndez C, Rogerson S, Dobaño C, Moncunill G. Chronic malaria exposure is associated with inhibitory markers on T cells that correlate with atypical memory and marginal zone-like B cells. Clin Exp Immunol 2024; 216:172-191. [PMID: 38387476 PMCID: PMC11036110 DOI: 10.1093/cei/uxae015] [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: 10/13/2023] [Revised: 12/05/2023] [Accepted: 02/22/2024] [Indexed: 02/24/2024] Open
Abstract
Chronic immune activation from persistent malaria infections can induce immunophenotypic changes associated with T-cell exhaustion. However, associations between T and B cells during chronic exposure remain undefined. We analyzed peripheral blood mononuclear cells from malaria-exposed pregnant women from Papua New Guinea and Spanish malaria-naïve individuals using flow cytometry to profile T-cell exhaustion markers phenotypically. T-cell lineage (CD3, CD4, and CD8), inhibitory (PD1, TIM3, LAG3, CTLA4, and 2B4), and senescence (CD28-) markers were assessed. Dimensionality reduction methods revealed increased PD1, TIM3, and LAG3 expression in malaria-exposed individuals. Manual gating confirmed significantly higher frequencies of PD1+CD4+ and CD4+, CD8+, and double-negative (DN) T cells expressing TIM3 in malaria-exposed individuals. Increased frequencies of T cells co-expressing multiple markers were also found in malaria-exposed individuals. T-cell data were analyzed with B-cell populations from a previous study where we reported an alteration of B-cell subsets, including increased frequencies of atypical memory B cells (aMBC) and reduction in marginal zone (MZ-like) B cells during malaria exposure. Frequencies of aMBC subsets and MZ-like B cells expressing CD95+ had significant positive correlations with CD28+PD1+TIM3+CD4+ and DN T cells and CD28+TIM3+2B4+CD8+ T cells. Frequencies of aMBC, known to associate with malaria anemia, were inversely correlated with hemoglobin levels in malaria-exposed women. Similarly, inverse correlations with hemoglobin levels were found for TIM3+CD8+ and CD28+PD1+TIM3+CD4+ T cells. Our findings provide further insights into the effects of chronic malaria exposure on circulating B- and T-cell populations, which could impact immunity and responses to vaccination.
Collapse
Affiliation(s)
- Robert A Mitchell
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Itziar Ubillos
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Pilar Requena
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Joseph J Campo
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Antigen Discovery Inc., Irvine, CA, USA
| | - Maria Ome-Kaius
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Sarah Hanieh
- University of Melbourne, Melbourne, VIC, Australia
| | - Alexandra Umbers
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Paula Samol
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Diana Barrios
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Azucena Bardají
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Ivo Mueller
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Clara Menéndez
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | | | - Carlota Dobaño
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| |
Collapse
|
34
|
Vargas Aguilar S, Cui M, Tan W, Sanchez-Ortiz E, Bassel-Duby R, Liu N, Olson EN. The PD-1-PD-L1 pathway maintains an immunosuppressive environment essential for neonatal heart regeneration. NATURE CARDIOVASCULAR RESEARCH 2024; 3:389-402. [PMID: 38737787 PMCID: PMC11086661 DOI: 10.1038/s44161-024-00447-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/05/2024] [Indexed: 05/14/2024]
Abstract
The adult mouse heart responds to injury by scarring with consequent loss of contractile function, whereas the neonatal heart possesses the ability to regenerate. Activation of the immune system is among the first events upon tissue injury. It has been shown that immune response kinetics differ between regeneration and pathological remodeling, yet the underlying mechanisms of the distinct immune reactions during tissue healing remain unclear. Here we show that the immunomodulatory PD-1-PD-L1 pathway is highly active in regenerative neonatal hearts but rapidly silenced later in life. Deletion of the PD-1 receptor or inactivation of its ligand PD-L1 prevented regeneration of neonatal hearts after injury. Disruption of the pathway during neonatal cardiac injury led to increased inflammation and aberrant T cell activation, which ultimately impaired cardiac regeneration. Our findings reveal an immunomodulatory and cardioprotective role for the PD-1-PD-L1 pathway in heart regeneration and offer potential avenues for the control of adult tissue regeneration.
Collapse
Affiliation(s)
- Stephanie Vargas Aguilar
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- These authors contributed equally: Stephanie Vargas Aguilar, Miao Cui
| | - Miao Cui
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Cardiology, Boston Children's Hospital and Department of Genetics, Harvard Medical School, Boston, MA, USA
- These authors contributed equally: Stephanie Vargas Aguilar, Miao Cui
| | - Wei Tan
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Efrain Sanchez-Ortiz
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ning Liu
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eric N Olson
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
35
|
Feng M, Zhang B, Li G, Yang Y, Liu J, Zhang Z, Zhou B, Zhang H. BACH2-mediated CD28 and CD40LG axes contribute to pathogenesis and progression of T-cell lymphoblastic leukemia. Cell Death Dis 2024; 15:59. [PMID: 38233409 PMCID: PMC10794190 DOI: 10.1038/s41419-024-06453-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/19/2024]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive subtype of ALL characterized by its high heterogeneity and unfavorable clinical features. Despite improved insights in genetic and epigenetic landscapes of T-ALL, the molecular mechanisms that drive malignant T-cell development remain unclear. BTB and CNC homology 2 (BACH2) is a lymphoid-specific transcription repressor recognized as a tumor suppressor in B-cell malignancies, but little is known about its function and regulatory network in T-ALL. Here we found extremely low levels of BACH2 in T-ALL clinical samples and cell lines compared to normal T cells. Overexpression of BACH2 in T-ALL cells not only induced cell growth retardation but also inhibited cancer progression and infiltration in xenografts. Further RNA sequencing (RNA-seq) analysis revealed significant alterations in regulation of defense and immune responses in T-ALL cells upon BACH2 overexpression. Strikingly, CD28 and CD40LG, two essential stimulatory molecules on T cells, were for the first time identified as novel downstream targets repressed by BACH2 in T-ALL cells. Interestingly, both CD28 and CD40LG were indispensable for T-ALL survival, since largely or completely silencing CD28 and CD40LG led to rapid cell death, whereas partial knockdown of them resulted in cell-cycle arrest and enhanced apoptosis. More importantly, BACH2-mediated CD28 and CD40LG signals contributed to cell migration and dissemination of T-ALL cells to the bone marrow, thus adding a new layer to the BACH2-mediated tumor immunoregulation in T-cell malignancies.
Collapse
Affiliation(s)
- Min Feng
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, 650118, China
| | - Bailing Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guilan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, 650118, China
| | - Yan Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, 650118, China
| | - Jiangyuan Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, 650118, China
| | - Ziting Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, 650118, China
| | - Bing Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Han Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, 650118, China.
| |
Collapse
|
36
|
Poddighe D, Dossybayeva K, Kozhakhmetov S, Rozenson R, Assylbekova M. Double-Negative T (DNT) Cells in Patients with Systemic Lupus Erythematosus. Biomedicines 2024; 12:166. [PMID: 38255272 PMCID: PMC10812956 DOI: 10.3390/biomedicines12010166] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Double-negative T (DNT) cells are a rare and unconventional T-lymphocyte subpopulation lacking both CD4 and CD8 markers. Their immunopathological roles and clinical relevance have yet to be elucidated. Beyond autoimmune lymphoproliferative syndrome (ALPS), these cells may also play a role in rheumatic disorders, including systemic lupus erythematosus (SLE); indeed, these two diseases share several autoimmune manifestations (including nephritis). Moreover, one of the main experimental murine models used to investigate lupus, namely the MRL/lpr mouse, is characterized by an expansion of DNT cells, which can support the production of pathogenic autoantibodies and/or modulate the immune response in this context. However, lupus murine models are not completely consistent with their human SLE counterpart, of course. In this mini review, we summarize and analyze the most relevant clinical studies investigating the DNT cell population in SLE patients. Overall, based on the present literature review and analysis, DNT cell homeostasis seems to be altered in patients with SLE. Indeed, most of the available clinical studies (which include both adults and children) reported an increased DNT cell percentage in SLE patients, especially during the active phases, even though no clear correlation with disease activity and/or inflammatory parameters has been clearly established. Well-designed, standardized, and longitudinal clinical studies focused on DNT cell population are needed, in order to further elucidate the actual contribution of these cells in SLE pathogenesis and their interactions with other immune cells (also implicated and/or altered in SLE, such as basophils), and clarify whether their expansion and/or immunophenotypic aspects may have any immunopathological relevance (and, then, represent potential disease markers and, in perspective, even therapeutic targets) or are just an unspecific epiphenomenon of autoimmunity.
Collapse
Affiliation(s)
- Dimitri Poddighe
- School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan;
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health, University Medical Center, Astana 010000, Kazakhstan;
| | | | - Samat Kozhakhmetov
- Center for Life Science, National Laboratory Astana, Astana 010000, Kazakhstan;
| | - Rafail Rozenson
- Department of Children’s Diseases n.1, Astana Medical University, Astana 010000, Kazakhstan;
| | - Maykesh Assylbekova
- Clinical Academic Department of Pediatrics, National Research Center for Maternal and Child Health, University Medical Center, Astana 010000, Kazakhstan;
| |
Collapse
|
37
|
Stein SR, Platt AP, Teague HL, Anthony SM, Reeder RJ, Cooper K, Byrum R, Drawbaugh DJ, Liu DX, Burdette TL, Hadley K, Barr B, Warner S, Rodriguez-Hernandez F, Johnson C, Stanek P, Hischak J, Kendall H, Huzella LM, Strich JR, Herbert R, St. Claire M, Vannella KM, Holbrook MR, Chertow DS. Clinical and Immunologic Correlates of Vasodilatory Shock Among Ebola Virus-Infected Nonhuman Primates in a Critical Care Model. J Infect Dis 2023; 228:S635-S647. [PMID: 37652048 PMCID: PMC10651209 DOI: 10.1093/infdis/jiad374] [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: 05/19/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Existing models of Ebola virus infection have not fully characterized the pathophysiology of shock in connection with daily virologic, clinical, and immunologic parameters. We implemented a nonhuman primate critical care model to investigate these associations. METHODS Two rhesus macaques received a target dose of 1000 plaque-forming units of Ebola virus intramuscularly with supportive care initiated on day 3. High-dimensional spectral cytometry was used to phenotype neutrophils and peripheral blood mononuclear cells daily. RESULTS We observed progressive vasodilatory shock with preserved cardiac function following viremia onset on day 5. Multiorgan dysfunction began on day 6 coincident with the nadir of circulating neutrophils. Consumptive coagulopathy and anemia occurred on days 7 to 8 along with irreversible shock, followed by death. The monocyte repertoire began shifting on day 4 with a decline in classical and expansion of double-negative monocytes. A selective loss of CXCR3-positive B and T cells, expansion of naive B cells, and activation of natural killer cells followed viremia onset. CONCLUSIONS Our model allows for high-fidelity characterization of the pathophysiology of acute Ebola virus infection with host innate and adaptive immune responses, which may advance host-targeted therapy design and evaluation for use after the onset of multiorgan failure.
Collapse
Affiliation(s)
- Sydney R Stein
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
| | - Andrew P Platt
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
| | - Heather L Teague
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
- Pathogenesis and Therapeutics Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| | - Scott M Anthony
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Rebecca J Reeder
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Kurt Cooper
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Russell Byrum
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - David J Drawbaugh
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - David X Liu
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Tracey L Burdette
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Kyra Hadley
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Bobbi Barr
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Seth Warner
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
- Pathogenesis and Therapeutics Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| | - Francisco Rodriguez-Hernandez
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Cristal Johnson
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Phil Stanek
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Joseph Hischak
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Heather Kendall
- Experimental Primate Virology Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Poolesville, Maryland, USA
| | - Louis M Huzella
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Jeffrey R Strich
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
- Pathogenesis and Therapeutics Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| | - Richard Herbert
- Experimental Primate Virology Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Poolesville, Maryland, USA
| | - Marisa St. Claire
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Kevin M Vannella
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
| | - Michael R Holbrook
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Daniel S Chertow
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
| |
Collapse
|
38
|
DuComb EA, Collins CC, Cupak D, Wagner S, Khan FB, Budd RC, Kinsey CM. Endobronchial ultrasound-guided transbronchial needle injection of cisplatin results in dynamic changes in the tumor immune microenvironment. Respir Med Res 2023; 84:100994. [PMID: 37307616 DOI: 10.1016/j.resmer.2023.100994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/14/2022] [Accepted: 12/26/2022] [Indexed: 01/12/2023]
Abstract
RATIONALE Direct intratumoral delivery of cisplatin via endobronchial ultrasound guided-transbronchial needle injections (EBUS-TBNI) is a novel approach for salvage treatment of advanced stage non-small cell lung cancer (NSCLC). The goal of this study was to evaluate changes in the tumor immune microenvironment during the course of EBUS-TBNI cisplatin therapy. METHODS Under an IRB approved protocol, patients with recurrence after radiation therapy who were not receiving other cytotoxic therapy, were prospectively enrolled, and underwent weekly treatments with EBUS-TBNI with additional biopsies obtained for research. Needle aspiration was performed prior to cisplatin delivery at each procedure. Samples were evaluated by flow cytometry for the presence of immune cell types. RESULTS Three of the six patients responded to the therapy based on RECIST criteria. Compared to the pre-treatment baseline, intratumoral neutrophils increased in 5 of the 6 patients (p = 0.041), with an average increase of 27.1%, but was not associated with response. A lower pre-treatment CD8+/CD4+ ratio at baseline was associated with response (P = 0.01). Responders demonstrated a lower final proportion of PD-1+ CD8+ T cells compared to non-responders (8.6% vs. 62.3%, respectively, P<0.001. Lower doses of intratumoral cisplatin were associated with subsequent increases in CD8+ T cells within the tumor microenvironment (P = 0.008). CONCLUSIONS EBUS-TBNI cisplatin resulted in significant alterations in the tumor immune microenvironment. Further studies are needed to determine if the changes seen here generalize to larger cohorts.
Collapse
Affiliation(s)
- Emily A DuComb
- Division of Pulmonary and Critical Care, University of Vermont College of Medicine, Burlington VT, United States
| | - Cheryl C Collins
- Division of Immunobiology, University of Vermont, University of Vermont College of Medicine, Burlington VT, United States
| | - Dolores Cupak
- Division of Immunobiology, University of Vermont, University of Vermont College of Medicine, Burlington VT, United States
| | - Sarah Wagner
- Division of Pulmonary and Critical Care, University of Vermont College of Medicine, Burlington VT, United States
| | - Farrah B Khan
- Division of Hematology and Oncology, University of Vermont College of Medicine, Burlington VT
| | - Ralph C Budd
- Division of Immunobiology, University of Vermont, University of Vermont College of Medicine, Burlington VT, United States
| | - C Matthew Kinsey
- Division of Pulmonary and Critical Care, University of Vermont College of Medicine, Burlington VT, United States.
| |
Collapse
|
39
|
Perrone R, Ashok Kumaar PV, Haky L, Hahn C, Riley R, Balough J, Zaza G, Soygur B, Hung K, Prado L, Kasler HG, Tiwari R, Matsui H, Hormazabal GV, Heckenbach I, Scheibye-Knudsen M, Duncan FE, Verdin E. CD38 regulates ovarian function and fecundity via NAD + metabolism. iScience 2023; 26:107949. [PMID: 37822499 PMCID: PMC10562803 DOI: 10.1016/j.isci.2023.107949] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/24/2023] [Accepted: 09/14/2023] [Indexed: 10/13/2023] Open
Abstract
Mammalian female reproductive lifespan is typically significantly shorter than life expectancy and is associated with a decrease in ovarian NAD+ levels. However, the mechanisms underlying this loss of ovarian NAD+ are unclear. Here, we show that CD38, an NAD+ consuming enzyme, is expressed in the ovarian extrafollicular space, primarily in immune cells, and its levels increase with reproductive age. Reproductively young mice lacking CD38 exhibit larger primordial follicle pools, elevated ovarian NAD+ levels, and increased fecundity relative to wild type controls. This larger ovarian reserve results from a prolonged window of follicle formation during early development. However, the beneficial effect of CD38 loss on reproductive function is not maintained at advanced age. Our results demonstrate a novel role of CD38 in regulating ovarian NAD+ metabolism and establishing the ovarian reserve, a critical process that dictates a female's reproductive lifespan.
Collapse
Affiliation(s)
| | | | - Lauren Haky
- Buck Institute for Research on Aging, Novato, CA, USA
- The Dominican University of California, San Rafael, CA, USA
| | - Cosmo Hahn
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Julia Balough
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Giuliana Zaza
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Bikem Soygur
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Kaitlyn Hung
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Leandro Prado
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Ritesh Tiwari
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | | | - Indra Heckenbach
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Francesca E. Duncan
- Buck Institute for Research on Aging, Novato, CA, USA
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| |
Collapse
|
40
|
Shi M, Wang Y, Zhang H, Ling Z, Chen X, Wang C, Liu J, Ma Y. Single-cell RNA sequencing shows the immune cell landscape in the kidneys of patients with idiopathic membranous nephropathy. Front Immunol 2023; 14:1203062. [PMID: 37731504 PMCID: PMC10507359 DOI: 10.3389/fimmu.2023.1203062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 08/18/2023] [Indexed: 09/22/2023] Open
Abstract
Idiopathic membranous nephropathy (IMN) is a leading pathological type of the adult primary nephrotic syndrome. Some patients develop end-stage renal disease due to poor response to treatment with steroid and immunosuppressive agents. In order to explore the molecular mechanism of IMN, we collected renal tissue samples from IMN patients and healthy controls and performed analysis by single-cell RNA sequencing (scRNA-seq). A total of 11 kidney cell clusters were identified, including multiple myeloid cell clusters, NK/T cell clusters, and B cell clusters. Most kidney parenchymal and immune cells were enriched in the regulation of immune response, inflammation, fibrosis and endoplasmic reticulum stress. The macrophage population in the IMN group showed a highly activated profile with up-regulated genes related to chemotaxis, inflammation, phagocytosis and fibrosis. CD8+ T cells continued to be cytotoxic in IMN; however, a transition to "inflammageing" GZMK+ CD8+ T cells was observed. The proportion of activated B cells in renal tissues of IMN patients was much higher than that of normal controls, indicating that B cells in IMN might be activated by constant antigenic stimulation. Moreover, the cell-cell interaction analysis revealed the potential communication between renal glomerular cells and immune cells in IMN. Overall, scRNA-seq was applied to IMN to unravel the characteristics of immune cells and elucidate possible underlying mechanisms involved in the pathogenesis of IMN.
Collapse
Affiliation(s)
- Manman Shi
- Department of Nephrology, Traditional Chinese Medicine Hospital of Kunshan, Kunshan, Jiangsu, China
| | - Yuxin Wang
- Department of Nephrology, Traditional Chinese Medicine Hospital of Kunshan, Kunshan, Jiangsu, China
| | - Huan Zhang
- Department of Nephrology, Shanghai Changhai Hospital, Navy Medical University, Shanghai, China
| | - Zicheng Ling
- Department of Internal Medicine, Weiting Community Health Center of Suzhou Industrial Park, Suzhou, Jiangsu, China
| | - Xue Chen
- Department of Nephrology, Traditional Chinese Medicine Hospital of Kunshan, Kunshan, Jiangsu, China
| | - Chaojun Wang
- Department of Nephrology, Traditional Chinese Medicine Hospital of Kunshan, Kunshan, Jiangsu, China
| | - Jian Liu
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yuhua Ma
- Department of Nephrology, Traditional Chinese Medicine Hospital of Kunshan, Kunshan, Jiangsu, China
| |
Collapse
|
41
|
Chen H, Xie X, Ma J, Fu L, Zhao X, Xing T, Gao C, Wu R, Chen Z. Elevated TCR-αβ + double-negative T cells in pediatric patients with acquired aplastic anemia. Clin Chim Acta 2023; 548:117492. [PMID: 37479012 DOI: 10.1016/j.cca.2023.117492] [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: 02/20/2023] [Revised: 06/19/2023] [Accepted: 07/16/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND AND AIMS The pathophysiology of acquired aplastic anemia (aAA) is most associated with T cell mediated immune dysfunction, but the role of CD4- CD8- double negative T cells (DNTs) in pediatric patients with aAA is unclear. In this study, we aimed to investigate the proportion of TCR-αβ+ DNTs in pediatric patients with aAA and correlation with the response to immunosuppressive therapy (IST). MATERIALS AND METHODS Assessment of DNTs from peripheral blood was done by sensitive multi-color flow cytometry. The potential clinical value of TCR-αβ+ DNTs was then assessed by the receiver operating characteristic (ROC) curves. RESULTS The retrospective study evaluated 164 pediatric patients with aAA and 105 healthy donors (HD). Our data showed higher proportion of TCR-αβ+ DNTs in total lymphocytes [1.04% (0.79%-1.40%) vs 0.69% (0.47%-0.87%), p < 0.001] and CD3+ T cells [1.52% (1.10%-1.96%) vs 1.10% (0.70%-1.40%), p < 0.001] in aAA compared to HD. Patients with SAA/VSAA achieving complete response (CR) after IST had a higher proportion of TCR-αβ+ DNTs at initial diagnosis, than those not achieving CR for total (1.21%±0.39 vs 0.78%±0.38, p < 0.05) and CD3+ T cells (1.74%±0.53 vs 1.15%±0.59, p < 0.05). The ROC analysis showed areas under the curves (AUCs) for TCR-αβ+ DNT proportion in lymphocytes and CD3+ T cells were 0.756 (cutoff value 1.33, p < 0.05) and 0.758 (cutoff value 1.38, p < 0.05), respectively. And the complete response rate was higher in TCR-αβ+ DNT proportion high group than in TCR-αβ+ DNT proportion low group at baseline (p < 0.001). CONCLUSION Our observations suggest that elevated TCR-αβ+ DNTs seems to play a role in the pathogenesis of aAA, and it was involve in immune response to IST.
Collapse
Affiliation(s)
- Hui Chen
- Hematologic Disease Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Xingjuan Xie
- Hematologic Disease Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Jie Ma
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Lingling Fu
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Xiaoxi Zhao
- Hematologic Disease Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Tianyu Xing
- Hematologic Disease Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Chao Gao
- Hematologic Disease Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Runhui Wu
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China.
| | - Zhenping Chen
- Hematologic Disease Laboratory, Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China.
| |
Collapse
|
42
|
Fenton KA, Pedersen HL. Advanced methods and novel biomarkers in autoimmune diseases ‑ a review of the recent years progress in systemic lupus erythematosus. Front Med (Lausanne) 2023; 10:1183535. [PMID: 37425332 PMCID: PMC10326284 DOI: 10.3389/fmed.2023.1183535] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/01/2023] [Indexed: 07/11/2023] Open
Abstract
There are several autoimmune and rheumatic diseases affecting different organs of the human body. Multiple sclerosis (MS) mainly affects brain, rheumatoid arthritis (RA) mainly affects joints, Type 1 diabetes (T1D) mainly affects pancreas, Sjogren's syndrome (SS) mainly affects salivary glands, while systemic lupus erythematosus (SLE) affects almost every organ of the body. Autoimmune diseases are characterized by production of autoantibodies, activation of immune cells, increased expression of pro-inflammatory cytokines, and activation of type I interferons. Despite improvements in treatments and diagnostic tools, the time it takes for the patients to be diagnosed is too long, and the main treatment for these diseases is still non-specific anti-inflammatory drugs. Thus, there is an urgent need for better biomarkers, as well as tailored, personalized treatment. This review focus on SLE and the organs affected in this disease. We have used the results from various rheumatic and autoimmune diseases and the organs involved with an aim to identify advanced methods and possible biomarkers to be utilized in the diagnosis of SLE, disease monitoring, and response to treatment.
Collapse
Affiliation(s)
- Kristin Andreassen Fenton
- UiT The Arctic University of Norway, Tromsø, Norway
- Centre of Clinical Research and Education, University Hospital of North Norway, Tromsø, Norway
| | - Hege Lynum Pedersen
- UiT The Arctic University of Norway, Tromsø, Norway
- Centre of Clinical Research and Education, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
43
|
Giacomelli M, Monti M, Pezzola DC, Lonardi S, Bugatti M, Missale F, Cioncada R, Melocchi L, Giustini V, Villanacci V, Baronchelli C, Manenti S, Imberti L, Giurisato E, Vermi W. Immuno-Contexture and Immune Checkpoint Molecule Expression in Mismatch Repair Proficient Colorectal Carcinoma. Cancers (Basel) 2023; 15:3097. [PMID: 37370706 DOI: 10.3390/cancers15123097] [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: 05/05/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Colorectal carcinoma (CRC) represents a lethal disease with heterogeneous outcomes. Only patients with mismatch repair (MMR) deficient CRC showing microsatellite instability and hyper-mutated tumors can obtain clinical benefits from current immune checkpoint blockades; on the other hand, immune- or target-based therapeutic strategies are very limited for subjects with mismatch repair proficient CRC (CRCpMMR). Here, we report a comprehensive typing of immune infiltrating cells in CRCpMMR. We also tested the expression and interferon-γ-modulation of PD-L1/CD274. Relevant findings were subsequently validated by immunohistochemistry on fixed materials. CRCpMMR contain a significantly increased fraction of CD163+ macrophages (TAMs) expressing TREM2 and CD66+ neutrophils (TANs) together with decrease in CD4-CD8-CD3+ double negative T lymphocytes (DNTs); no differences were revealed by the analysis of conventional and plasmacytoid dendritic cell populations. A fraction of tumor-infiltrating T-cells displays an exhausted phenotype, co-expressing PD-1 and TIM-3. Remarkably, expression of PD-L1 on fresh tumor cells and TAMs was undetectable even after in vitro stimulation with interferon-γ. These findings confirm the immune suppressive microenvironment of CRCpMMR characterized by dense infiltration of TAMs, occurrence of TANs, lack of DNTs, T-cell exhaustion, and interferon-γ unresponsiveness by host and tumor cells. Appropriate bypass strategies should consider these combinations of immune escape mechanisms in CRCpMMR.
Collapse
Affiliation(s)
- Mauro Giacomelli
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Diego Cesare Pezzola
- Department of Surgery, Surgery Division II, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Mattia Bugatti
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Francesco Missale
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Department of Head & Neck Oncology & Surgery Otorhinolaryngology, Antoni Van Leeuwenhoek-Nederlands Kanker Instituut, 1066 CX Amsterdam, The Netherlands
| | - Rossella Cioncada
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Laura Melocchi
- Department of Pathology, Fondazione Poliambulanza, 25124 Brescia, Italy
| | - Viviana Giustini
- CREA Laboratory, AIL Center for Hemato-Oncologic Research, Diagnostic Department, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Vincenzo Villanacci
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Carla Baronchelli
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Stefania Manenti
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Luisa Imberti
- Section of Microbiology, University of Brescia, 25123 Brescia, Italy
| | - Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| | - William Vermi
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Department of Pathology and Immunology, School of Medicine, Washington University, Saint Louis, MO 63130, USA
| |
Collapse
|
44
|
Chamorro BM, Luca KD, Swaminathan G, Rochereau N, Majorel J, Poulet H, Chanut B, Piney L, Mundt E, Paul S. Mucosal Vaccination with Live Attenuated Bordetella bronchiseptica Protects against Challenge in Wistar Rats. Vaccines (Basel) 2023; 11:vaccines11050982. [PMID: 37243086 DOI: 10.3390/vaccines11050982] [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: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Bordetella bronchiseptica (Bb) is a Gram-negative bacterium responsible for canine infectious respiratory disease complex (CIRDC). Several vaccines targeting this pathogen are currently licensed for use in dogs, but their mechanism of action and the correlates of protection are not fully understood. To investigate this, we used a rat model to examine the immune responses induced and the protection conferred by a canine mucosal vaccine after challenge. Wistar rats were vaccinated orally or intranasally on D0 and D21 with a live attenuated Bb vaccine strain. At D35, the rats of all groups were inoculated with 103 CFU of a pathogenic strain of B. bronchiseptica. Animals vaccinated via either the intranasal or the oral route had Bb-specific IgG and IgM in their serum and Bb-specific IgA in nasal lavages. Bacterial load in the trachea, lung, and nasal lavages was lower in vaccinated animals than in non-vaccinated control animals. Interestingly, coughing improved in the group vaccinated intranasally, but not in the orally vaccinated or control group. These results suggest that mucosal vaccination can induce mucosal immune responses and provide protection against a Bb challenge. This study also highlights the advantages of a rat model as a tool for studying candidate vaccines and routes of administration for dogs.
Collapse
Affiliation(s)
- Beatriz Miguelena Chamorro
- CIRI-Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, F69007 Lyon, France
- Boehringer Ingelheim, Global Innovation, F69800 Saint Priest, France
| | - Karelle De Luca
- Boehringer Ingelheim, Global Innovation, F69800 Saint Priest, France
| | - Gokul Swaminathan
- Boehringer Ingelheim, Global Innovation, F69800 Saint Priest, France
| | - Nicolas Rochereau
- CIRI-Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, F69007 Lyon, France
| | - Jade Majorel
- CIRI-Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, F69007 Lyon, France
| | - Hervé Poulet
- Boehringer Ingelheim, Global Innovation, F69800 Saint Priest, France
| | - Blandine Chanut
- CIRI-Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, F69007 Lyon, France
| | - Lauriane Piney
- Boehringer Ingelheim, Global Innovation, F69800 Saint Priest, France
| | - Egbert Mundt
- Boehringer Ingelheim, Global Innovation, F69800 Saint Priest, France
| | - Stéphane Paul
- CIRI-Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, F69007 Lyon, France
- 3CIC Inserm 1408 Vaccinology, F42023 Saint-Etienne, France
| |
Collapse
|
45
|
Koh CC, Neves EGA, de Souza-Silva TG, Carvalho AC, Pinto CHR, Sobreira Galdino A, Gollob KJ, Dutra WO. Cytokine Networks as Targets for Preventing and Controlling Chagas Heart Disease. Pathogens 2023; 12:171. [PMID: 36839443 PMCID: PMC9966322 DOI: 10.3390/pathogens12020171] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Chagas disease, a neglected disease caused by the protozoan Trypanosoma cruzi, is endemic in 21 Latin American countries, affecting 6-8 million people. Increasing numbers of Chagas disease cases have also been reported in non-endemic countries due to migration, contamination via blood transfusions or organ transplantation, characterizing Chagas as an emerging disease in such regions. While most individuals in the chronic phase of Chagas disease remain in an asymptomatic clinical form named indeterminate, approximately 30% of the patients develop a cardiomyopathy that is amongst the deadliest cardiopathies known. The clinical distinctions between the indeterminate and the cardiac clinical forms are associated with different immune responses mediated by innate and adaptive cells. In this review, we present a collection of studies focusing on the human disease, discussing several aspects that demonstrate the association between chemokines, cytokines, and cytotoxic molecules with the distinct clinical outcomes of human infection with Trypanosoma cruzi. In addition, we discuss the role of gene polymorphisms in the transcriptional control of these immunoregulatory molecules. Finally, we discuss the potential application of cytokine expression and gene polymorphisms as markers of susceptibility to developing the severe form of Chagas disease, and as targets for disease control.
Collapse
Affiliation(s)
- Carolina Cattoni Koh
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Eula G. A. Neves
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Thaiany Goulart de Souza-Silva
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Ana Carolina Carvalho
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Cecília Horta Ramalho Pinto
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Alexsandro Sobreira Galdino
- Laboratório de Biotecnologia de Microrganismos, Universidade Federal de São João Del-Rei (UFSJ), Campus Centro Oeste, Divinópolis 35501-296, MG, Brazil
| | - Kenneth J. Gollob
- Hospital Israelita Albert Einstein, São Paulo 05652-900, SP, Brazil
- Instituto Nacional de Ciências e Tecnologia em Doenças Tropicais, INCT-DT, Salvador 40110-160, BA, Brazil
| | - Walderez Ornelas Dutra
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
- Instituto Nacional de Ciências e Tecnologia em Doenças Tropicais, INCT-DT, Salvador 40110-160, BA, Brazil
| |
Collapse
|
46
|
Kopitar AN, Repas J, Janžič L, Bizjak M, Vesel TT, Emeršič N, Avramovič MZ, Ihan A, Avčin T, Pavlin M. Alterations in immunophenotype and metabolic profile of mononuclear cells during follow up in children with multisystem inflammatory syndrome (MIS-C). Front Immunol 2023; 14:1157702. [PMID: 37153551 PMCID: PMC10157053 DOI: 10.3389/fimmu.2023.1157702] [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: 02/02/2023] [Accepted: 03/29/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Although children seem to be less susceptible to COVID-19, some of them develop a rare but serious hyperinflammatory condition called multisystem inflammatory syndrome in children (MIS-C). While several studies describe the clinical conditions of acute MIS-C, the status of convalescent patients in the months after acute MIS-C is still unclear, especially the question of persistence of changes in the specific subpopulations of immune cells in the convalescent phase of the disease. Methods We therefore analyzed peripheral blood of 14 children with MIS-C at the onset of the disease (acute phase) and 2 to 6 months after disease onset (post-acute convalescent phase) for lymphocyte subsets and antigen-presenting cell (APC) phenotype. The results were compared with six healthy age-matched controls. Results All major lymphocyte populations (B cells, CD4 + and CD8+ T cells, and NK cells) were decreased in the acute phase and normalized in the convalescent phase. T cell activation was increased in the acute phase, followed by an increased proportion of γ/δ-double-negative T cells (γ/δ DN Ts) in the convalescent phase. B cell differentiation was impaired in the acute phase with a decreased proportion of CD21 expressing, activated/memory, and class-switched memory B cells, which normalized in the convalescent phase. The proportion of plasmacytoid dendritic cells, conventional type 2 dendritic cells, and classical monocytes were decreased, while the proportion of conventional type 1 dendritic cells was increased in the acute phase. Importantly the population of plasmacytoid dendritic cells remained decreased in the convalescent phase, while other APC populations normalized. Immunometabolic analysis of peripheral blood mononuclear cells (PBMCs) in the convalescent MIS-C showed comparable mitochondrial respiration and glycolysis rates to healthy controls. Conclusions While both immunophenotyping and immunometabolic analyzes showed that immune cells in the convalescent MIS-C phase normalized in many parameters, we found lower percentage of plasmablasts, lower expression of T cell co-receptors (CD3, CD4, and CD8), an increased percentage of γ/δ DN Ts and increased metabolic activity of CD3/CD28-stimulated T cells. Overall, the results suggest that inflammation persists for months after the onset of MIS-C, with significant alterations in some immune system parameters, which may also impair immune defense against viral infections.
Collapse
Affiliation(s)
- Andreja Nataša Kopitar
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jernej Repas
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Larisa Janžič
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Maša Bizjak
- Department for Allergology, Rheumatology and Clinical Immunology, Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Tina Tajnšek Vesel
- Department for Allergology, Rheumatology and Clinical Immunology, Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Nina Emeršič
- Department for Allergology, Rheumatology and Clinical Immunology, Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Mojca Zajc Avramovič
- Department for Allergology, Rheumatology and Clinical Immunology, Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Alojz Ihan
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tadej Avčin
- Department for Allergology, Rheumatology and Clinical Immunology, Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, Department of Pediatrics, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Tadej Avčin, ; Mojca Pavlin,
| | - Mojca Pavlin
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Tadej Avčin, ; Mojca Pavlin,
| |
Collapse
|
47
|
Xu W, Wan S, Xie B, Song X. Novel potential therapeutic targets of alopecia areata. Front Immunol 2023; 14:1148359. [PMID: 37153617 PMCID: PMC10154608 DOI: 10.3389/fimmu.2023.1148359] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/05/2023] [Indexed: 05/10/2023] Open
Abstract
Alopecia areata (AA) is a non-scarring hair loss disorder caused by autoimmunity. The immune collapse of the hair follicle, where interferon-gamma (IFN-γ) and CD8+ T cells accumulate, is a key factor in AA. However, the exact functional mechanism remains unclear. Therefore, AA treatment has poor efficacy maintenance and high relapse rate after drug withdrawal. Recent studies show that immune-related cells and molecules affect AA. These cells communicate through autocrine and paracrine signals. Various cytokines, chemokines and growth factors mediate this crosstalk. In addition, adipose-derived stem cells (ADSCs), gut microbiota, hair follicle melanocytes, non-coding RNAs and specific regulatory factors have crucial roles in intercellular communication without a clear cause, suggesting potential new targets for AA therapy. This review discusses the latest research on the possible pathogenesis and therapeutic targets of AA.
Collapse
Affiliation(s)
- Wen Xu
- School of Medicine, Zhejiang University, Hangzhou, China
- Department of Dermatology, Hangzhou Third People’s Hospital, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Sheng Wan
- Department of Dermatology, Hangzhou Third People’s Hospital, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo Xie
- Department of Dermatology, Hangzhou Third People’s Hospital, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiuzu Song
- Department of Dermatology, Hangzhou Third People’s Hospital, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Xiuzu Song,
| |
Collapse
|
48
|
Adoptive Cell Therapy for T-Cell Malignancies. Cancers (Basel) 2022; 15:cancers15010094. [PMID: 36612092 PMCID: PMC9817702 DOI: 10.3390/cancers15010094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
T-cell malignancies are often aggressive and associated with poor prognoses. Adoptive cell therapy has recently shown promise as a new line of therapy for patients with hematological malignancies. However, there are currently challenges in applying adoptive cell therapy to T-cell malignancies. Various approaches have been examined in preclinical and clinical studies to overcome these obstacles. This review aims to provide an overview of the recent progress on adoptive cell therapy for T-cell malignancies. The benefits and drawbacks of different types of adoptive cell therapy are discussed. The potential advantages and current applications of innate immune cell-based adoptive cell therapy for T cell malignancies are emphasized.
Collapse
|
49
|
Wang M, Wei Y, Li Y, Li H, Jin J, Lu Y, Li Q. Targeting breast cancer with a combination of DNT and LAG3 checkpoint blockage and its mechanism. Immun Inflamm Dis 2022; 10:e626. [PMID: 35894707 PMCID: PMC9274802 DOI: 10.1002/iid3.626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 11/06/2022] Open
Abstract
INTRODUCTION The characteristics of the tumor immune microenvironment (TIME) are closely related to immunotherapy. Breast cancer can benefit from immunotherapy, and its TIME is still unclear. METHODS We utilized mass cytometry to explore the immune cell heterogeneity in breast cancer. Double-negative T cells (DNTs) from healthy volunteers (HBs) were enriched in vitro. Flow cytometry was used to detect the cell surface receptors of cancer cells and DNT cells. The correlation between immune checkpoints and the abundance of immune cells or prognosis of breast cancer was analyzed by the TCGA database. The messenger RNA (mRNA) expression of functional genes was performed by quantitative real-time PCR. RESULTS We found that the frequencies of Granzyme B (GZMB)+ CD8+ T and GZMB+ DNT cells in cancer tissues (CA) of breast cancer were lower than those in blood samples of patients (PB), and the frequencies of programmed cell death protein 1 (PD1)+ CD8+ T and PD1+ DNT cells in CA were higher than those in PB. DNTs from HBs had a cytotoxic effect on MDA-MB-231. LAG3Ab could upregulate the mRNA expression of interferon gamma and perforin by increasing T-BET transcription to enhance the cytotoxicity of DNT cells in vitro. CONCLUSION Our study revealed the suppressive status of TIME in breast cancer and supported DNT cells had the potential to be applied as a novel adoptive cell therapy for TNBC either alone or combined with LAG3Ab.
Collapse
Affiliation(s)
- Miao Wang
- Department of OncologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Yuhan Wei
- Department of OncologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Yingrui Li
- Department of OncologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Hongzhong Li
- Chongqing Key Laboratory of Molecular Oncology and EpigeneticsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Jiangtao Jin
- Department of Intervention TherapyZezhou People's HospitalJinchengChina
| | - Yuting Lu
- Department of OncologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Qin Li
- Department of OncologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|