1
|
Kim JG, Kim M, Hong BK, Choe YH, Kim JR, Lee N, You S, Lee SI, Kim WU. Circulatory age-associated B cells: Their distinct transcriptomic characteristics and clinical significance in drug-naïve patients with rheumatoid arthritis. Clin Immunol 2025; 271:110425. [PMID: 39746429 DOI: 10.1016/j.clim.2024.110425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/02/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Age-associated B cells (ABCs) have been implicated in the pathogenesis of autoimmune diseases. However, the global gene expression and clinical significance of circulatory ABCs in rheumatoid arthritis (RA) remain poorly understood. Here, single-cell RNA sequencing identified nine B cell subsets in peripheral blood of RA patients, including ABCs. Increased phagocytosis and antigen presentation were functionally enriched by the genes expressed differentially in ABCs. Network analysis and in vitro experiments demonstrated SYK as a key regulator defining the myeloid-like phenotypes in ABCs. Flow cytometry showed that the proportion of ABCs correlated with RA activity and serum tumor necrosis factor-alpha level. Notably, ABCs above a cutoff threshold specifically distinguished RA from healthy controls and indicated higher disease activity. This study highlights the myeloid characteristics of circulatory ABCs regulated by SYK in RA. Increased ABCs may reflect disease activity and could serve as a potential biomarker in RA.
Collapse
Affiliation(s)
- Jung Gon Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Inje University Ilsan Paik Hospital, Goyang, Republic of Korea
| | - Mingyo Kim
- Division of Rheumatology, Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Bong-Ki Hong
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yong-Ho Choe
- Division of Rheumatology, Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Ju-Ryoung Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea
| | - Naeun Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sungyong You
- Urology and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sang-Il Lee
- Division of Rheumatology, Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, Republic of Korea.
| | - Wan-Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Younis DA, Marosvari M, Liu W, Pulikkot S, Cao Z, Zhou B, Vella AT, McArdle S, Hu L, Chen Y, Gan W, Yu J, Bruscia EM, Fan Z. CFTR dictates monocyte adhesion by facilitating integrin clustering but not activation. Proc Natl Acad Sci U S A 2025; 122:e2412717122. [PMID: 39813254 PMCID: PMC11760921 DOI: 10.1073/pnas.2412717122] [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: 06/24/2024] [Accepted: 12/21/2024] [Indexed: 01/18/2025] Open
Abstract
Monocytes are critical in controlling tissue infections and inflammation. Monocyte dysfunction contributes to the inflammatory pathogenesis of cystic fibrosis (CF) caused by CF transmembrane conductance regulator (CFTR) mutations, making CF a clinically relevant disease model for studying the contribution of monocytes to inflammation. Although CF monocytes exhibited adhesion defects, the precise mechanism is unclear. Herein, superresolution microscopy showed that an integrin clustering but not an integrin activation defect determines the adhesion defect in CFTR-deficient monocytes, challenging the existing paradigm emphasizing an integrin activation defect in CF patient monocytes. We further found that the clustering defect is accompanied by defects in CORO1A membrane recruitment, actin cortex formation, and CORO1A engagement with integrins. Complementing canonical studies of leukocyte adhesion focusing on integrin activation, we highlight the importance of integrin clustering in cell adhesion and report that integrin clustering and activation are distinctly regulated, warranting further investigation for selective targeting in therapeutic strategy design involving leukocyte-dependent inflammation.
Collapse
Affiliation(s)
| | - Mason Marosvari
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT06030
| | - Wei Liu
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT06030
| | - Sunitha Pulikkot
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT06030
| | - Ziming Cao
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT06030
| | - Beiyan Zhou
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT06030
| | - Anthony T. Vella
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT06030
| | - Sara McArdle
- Microscopy Core Facility, La Jolla Institute for Immunology, San Diego, CA92037
| | - Liang Hu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai201203, China
| | - Yunfeng Chen
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX77555
- Department of Pathology, University of Texas Medical Branch, Galveston, TX77555
| | - Wenqi Gan
- Department of Public Health Sciences, School of Medicine, UConn Health, Farmington, CT06030
| | - Ji Yu
- Center for Cell Analysis and Modeling, UConn Health, Farmington, CT06030
| | - Emanuela M. Bruscia
- Department of Pediatrics, School of Medicine, Yale University, New Haven, CT06510
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT06030
| |
Collapse
|
3
|
Pierini S, Gabbasov R, Oliveira-Nunes MC, Qureshi R, Worth A, Huang S, Nagar K, Griffin C, Lian L, Yashiro-Ohtani Y, Ross K, Sloas C, Ball M, Schott B, Sonawane P, Cornell L, Blumenthal D, Chhum S, Minutolo N, Ciccaglione K, Shaw L, Zentner I, Levitsky H, Shestova O, Gill S, Varghese B, Cushing D, Ceeraz DeLong S, Abramson S, Condamine T, Klichinsky M. Chimeric antigen receptor macrophages (CAR-M) sensitize HER2+ solid tumors to PD1 blockade in pre-clinical models. Nat Commun 2025; 16:706. [PMID: 39814734 PMCID: PMC11735936 DOI: 10.1038/s41467-024-55770-1] [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: 10/24/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025] Open
Abstract
We previously developed human CAR macrophages (CAR-M) and demonstrated redirection of macrophage anti-tumor function leading to tumor control in immunodeficient xenograft models. Here, we develop clinically relevant fully immunocompetent syngeneic models to evaluate the potential for CAR-M to remodel the tumor microenvironment (TME), induce T cell anti-tumor immunity, and sensitize solid tumors to PD1/PDL1 checkpoint inhibition. In vivo, anti-HER2 CAR-M significantly reduce tumor burden, prolong survival, remodel the TME, increase intratumoral T cell and natural killer (NK) cell infiltration, and induce antigen spreading. CAR-M therapy protects against antigen-negative relapses in a T cell dependent fashion, confirming long-term anti-tumor immunity. In HER2+ solid tumors with limited sensitivity to anti-PD1 (aPD1) monotherapy, the combination of CAR-M and aPD1 significantly improves tumor growth control, survival, and remodeling of the TME in pre-clinical models. These results demonstrate synergy between CAR-M and T cell checkpoint blockade and provide a strategy to potentially enhance response to aPD1 therapy for patients with non-responsive tumors.
Collapse
Affiliation(s)
| | | | | | | | | | - Shuo Huang
- Carisma Therapeutics Inc, Philadelphia, PA, USA
| | - Karan Nagar
- Carisma Therapeutics Inc, Philadelphia, PA, USA
| | | | - Lurong Lian
- Carisma Therapeutics Inc, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Lauren Shaw
- Carisma Therapeutics Inc, Philadelphia, PA, USA
| | | | | | - Olga Shestova
- Center for Cellular Immunotherapies, Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Saar Gill
- Center for Cellular Immunotherapies, Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
4
|
Liu J, Guo Q, Liu G, Wang W, Jin X, Hao B, Lei B. Immune pathogenic response landscape of acute posterior multifocal placoid pigment epitheliopathy revealed by scRNA sequencing. Genes Immun 2025:10.1038/s41435-024-00316-0. [PMID: 39774261 DOI: 10.1038/s41435-024-00316-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025]
Abstract
Acute posterior multifocal placoid pigment epitheliopathy (APMPPE) is an exceptionally rare inflammatory disorder affecting choroid and retinal pigment epithelial (RPE) cells. Although recent studies suggest an immune-driven nature, the underlying etiology of APMPPE remains elusive. In this study, we conducted a comprehensive investigation on the peripheral blood mononuclear cells (PBMCs) profile of an APMPPE patient using single-cell RNA sequencing. Our analysis revealed striking transcriptional alterations in monocytes within the PBMCs, identifying five distinct subpopulations: S100A12, CD16, pro-inflammatory, megakaryocyte-like, and NK-like monocyte subsets. Employing pseudotime inference, we observed a shift in APMPPE monocytes towards differentiation into inflammation-associated pro-inflammatory monocytes and a CD16 monocyte trajectory. Furthermore, we identified IFITM3 as a key player in the immune response driving the pathogenesis of APMPPE. Notably, two disease-relevant subgroups of monocytes, pro-inflammatory and CD16 monocytes, were implicated in APMPPE. CD16 monocytes, in particular, were involved in melanogenesis, suggesting that the abnormal expression of melanin in monocytes might result from autoimmune responses against pigment-enriched RPE cells. This study provided a comprehensive view of immune landscape in APMPPE, shedding light on the previously unrecognized contributions of pro-inflammatory and CD16 monocytes to this autoimmune condition.
Collapse
Affiliation(s)
- Jingyang Liu
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Qingge Guo
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Guangming Liu
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Weiping Wang
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Xiuxiu Jin
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China.
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China.
| | - Bingtao Hao
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China.
- Department of Immunology, School of Basic Medical, Zhengzhou University, Zhengzhou, Henan, China.
| | - Bo Lei
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China.
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China.
| |
Collapse
|
5
|
Stinson JA, Sheen A, Lax BM, Yang GN, Duhamel L, Santollani L, Fink E, Palmeri J, Wittrup KD. Tumor Integrin-Targeted Glucose Oxidase Enzyme Promotes ROS-Mediated Cell Death that Combines with Interferon Alpha Therapy for Tumor Control. Mol Cancer Ther 2025; 24:118-130. [PMID: 39382078 PMCID: PMC11695183 DOI: 10.1158/1535-7163.mct-24-0163] [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: 03/06/2024] [Revised: 07/14/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024]
Abstract
Although heightened intratumoral levels of reactive oxygen species (ROS) are typically associated with a suppressive tumor microenvironment, under certain conditions ROS contribute to tumor elimination. Treatment approaches, including some chemotherapy and radiation protocols, increase cancer cell ROS levels that influence their mechanism of cell death and subsequent recognition by the immune system. Furthermore, activated myeloid cells rapidly generate ROS upon encounter with pathogens or infected cells to eliminate disease, and recently, this effector function has been noted in cancer contexts as well. Collectively, ROS-induced cancer cell death may help initiate adaptive antitumor immune responses that could synergize with current approved immunotherapies, for improved control of solid tumors. In this work, we explore the use of glucose oxidase, an enzyme which produces hydrogen peroxide, a type of ROS, to therapeutically mimic the endogenous oxidative burst from myeloid cells to promote antigen generation within the tumor microenvironment. We engineer the enzyme to target pan-tumor-expressed integrins both as a tumor-agnostic therapeutic approach and as a strategy to prolong local enzyme activity following intratumoral administration. We found the targeted enzyme potently induced cancer cell death and enhanced cross-presentation by dendritic cells in vitro and further combined with interferon alpha for long-term tumor control in murine MC38 tumors in vivo. Optimizing the single-dose administration of this enzyme overcomes limitations with immunogenicity noted for other prooxidant enzyme approaches. Overall, our results suggest ROS-induced cell death can be harnessed for tumor control and highlight the potential use of designed enzyme therapies alongside immunotherapy against cancer.
Collapse
Affiliation(s)
- Jordan A. Stinson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Allison Sheen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Brianna M. Lax
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Grace N. Yang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Lauren Duhamel
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Luciano Santollani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Elizabeth Fink
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Joseph Palmeri
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - K. Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
6
|
Zheng F, Dong T, Chen Y, Wang L, Peng G. Border-associated macrophages: From physiology to therapeutic targets in Alzheimer's disease. Exp Neurol 2025; 383:115021. [PMID: 39461707 DOI: 10.1016/j.expneurol.2024.115021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
Border-associated macrophages (BAMs) constitute a highly heterogeneous group of central nervous system-resident macrophages at the brain boundaries. Despite their significance, BAMs have mainly been overlooked compared to microglia, resulting in a limited understanding of their functions. However, recent advancements in single-cell immunophenotyping and transcriptomic analyses of BAMs have revealed a previously unrecognized complexity in these cells, in addition to their critical roles under non-pathological conditions and diseases like Alzheimer's disease (AD), Parkinson's disease, glioma, and ischemic stroke. In this review, we discuss the origins, self-renewal capabilities, and extensive heterogeneity of BAMs, and clarify their important physiological functions such as immune monitoring, waste removal and vascular permeability regulation. We also summarize experimental evidence linking BAMs to the progression of AD. Finally, we review therapeutic strategies targeting brain innate immune cells mainly focusing on strategies aimed at modulating BAMs to treat AD and evaluate their potential in clinical applications.
Collapse
Affiliation(s)
- Fangxue Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Taiwei Dong
- Hangzhou Normal University School of Basic Medical Sciences, Hangzhou, China
| | - Yi Chen
- Department of Neurology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lang Wang
- Department of Neurology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, China.
| | - Guoping Peng
- Department of Neurology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
7
|
Bao C, Ma Q, Ying X, Wang F, Hou Y, Wang D, Zhu L, Huang J, He C. Histone lactylation in macrophage biology and disease: from plasticity regulation to therapeutic implications. EBioMedicine 2025; 111:105502. [PMID: 39662177 PMCID: PMC11697715 DOI: 10.1016/j.ebiom.2024.105502] [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: 07/30/2024] [Revised: 11/10/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024] Open
Abstract
Epigenetic modifications have been identified as critical molecular determinants influencing macrophage plasticity and heterogeneity. Among these, histone lactylation is a recently discovered epigenetic modification. Research examining the effects of histone lactylation on macrophage activation and polarization has grown substantially in recent years. Evidence increasingly suggests that lactate-mediated changes in histone lactylation levels within macrophages can modulate gene transcription, thereby contributing to the pathogenesis of various diseases. This review provides a comprehensive analysis of the role of histone lactylation in macrophage activation, exploring its discovery, effects, and association with macrophage diversity and phenotypic variability. Moreover, it highlights the impact of alterations in macrophage histone lactylation in diverse pathological contexts, such as inflammation, tumorigenesis, neurological disorders, and other complex conditions, and demonstrates the therapeutic potential of drugs targeting these epigenetic modifications. This mechanistic understanding provides insights into the underlying disease mechanisms and opens new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Chuncha Bao
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Qing Ma
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xihong Ying
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Fengsheng Wang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, PR China
| | - Yue Hou
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Dun Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Linsen Zhu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Jiapeng Huang
- Clinical Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, PR China.
| | - Chengqi He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, PR China.
| |
Collapse
|
8
|
de Castro Sampaio SS, Ramalho MCC, de Souza CS, de Almeida Rodrigues B, de Mendonça GRS, Lazarini M. RHO subfamily of small GTPases in the development and function of hematopoietic cells. J Cell Physiol 2025; 240:e31469. [PMID: 39434451 DOI: 10.1002/jcp.31469] [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: 07/25/2024] [Revised: 09/16/2024] [Accepted: 10/03/2024] [Indexed: 10/23/2024]
Abstract
RHOA, RHOB, and RHOC comprise a subfamily of RHO GTPase proteins famed for controlling cytoskeletal dynamics. RHO proteins operate downstream of multiple signals emerging from the microenvironment, leading to diverse cell responses, such as proliferation, adhesion, and migration. Therefore, RHO signaling has been centrally placed in the regulation of blood cells. Despite their high homology, unique roles of RHOA, RHOB, and RHOC have been described in hematopoietic cells. In this article, we overview the contribution of RHO proteins in the development and function of each blood cell lineage. Additionally, we highlight the aberrations of the RHO signaling pathways found in hematological malignancies, providing clues for the identification of new therapeutic targets.
Collapse
Affiliation(s)
| | | | - Caroline Santos de Souza
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Mariana Lazarini
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
9
|
Wang H, Li W, Lai Q, Huang Q, Ding H, Deng Z. Inflammatory Markers and Risk of Parkinson's Disease: A Population-Based Analysis. PARKINSON'S DISEASE 2024; 2024:4192853. [PMID: 39780847 PMCID: PMC11707066 DOI: 10.1155/padi/4192853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 11/05/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
Objected: Parkinson's disease (PD) is an important cause of neurological dysfunction, and the aim of this study was to explore whether neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), lymphocyte-to-monocyte ratio (LMR), systemic inflammatory response (SIRI), and systemic immune inflammation (SII) are associated with the risk of developing PD. Based on this, we may identify people at high risk for PD and intervene early. Method: Our study included 31,480 participants from the National Health and Nutrition Examination Survey (NHANES) between 2001 and 2018. Basic information and inflammation-related indicators were obtained by questionnaires and laboratory tests, respectively. NLR, PLR, LMR, SIRI, SII, and PD risk were analyzed using weighted logistic regression models. Results: There were 261 and 31,219 in the PD and non-PD groups, respectively, and the prevalence of PD was 0.83%. Separate analyses of NLR and PLR were conducted after fully adjusting for confounding factors. According to our analysis, there was an increased risk of PD for both NLR and PLR in the higher level group (Q4) as compared with the lower level group (Q1) (OR = 1.83 and 95% confidence interval (CI) = 1.09-3.07, and OR = 1.92 and 95% CI = 1.20-3.08). However, we did not find similar relationships in LMR, SIRI, and SII. Conclusions: There was a significant association between elevated levels of NLR, PLR, and PD risk, while LMR, SIRI, and SII were not statistically significant. It suggests that NLR or PLR could be used to screen people at risk of PD at an early stage. It is essential to conduct more large-scale prospective studies to investigate the role that NLR and PLR play in PD.
Collapse
Affiliation(s)
- Hongping Wang
- Department of Neurosurgery, Zigong Fourth People's Hospital, 19 Tanmulin Street, Zigong, Sichuan, China
| | - Wenqiang Li
- Department of Pulmonary and Critical Care Medicine, Zigong First People's Hospital, 42 Shangyihao Yizhi Street, Zigong, Sichuan, China
| | - Qun Lai
- Department of Hematology, Rheumatology and Immunology, Zigong Fourth People's Hospital, 19 Tanmulin Street, Zigong, Sichuan, China
| | - Qian Huang
- Department of Pulmonary and Critical Care Medicine, Dazhou Third People's Hospital, Dazhou, Sichuan, China
| | - Hao Ding
- Department of Neurosurgery, Zigong Fourth People's Hospital, 19 Tanmulin Street, Zigong, Sichuan, China
| | - Zhiping Deng
- Department of Pulmonary and Critical Care Medicine, Zigong First People's Hospital, 42 Shangyihao Yizhi Street, Zigong, Sichuan, China
| |
Collapse
|
10
|
Lin F, Yin S, Zhang Z, Yu Y, Fang H, Liang Z, Zhu R, Zhou H, Li J, Cao K, Guo W, Qin S, Zhang Y, Lu C, Li H, Liu S, Zhang H, Ye B, Lin J, Li Y, Kang X, Xi JJ, Chen PR. Multimodal targeting chimeras enable integrated immunotherapy leveraging tumor-immune microenvironment. Cell 2024; 187:7470-7491.e32. [PMID: 39504957 DOI: 10.1016/j.cell.2024.10.016] [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: 07/13/2022] [Revised: 08/02/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024]
Abstract
Although immunotherapy has revolutionized cancer treatment, its efficacy is affected by multiple factors, particularly those derived from the complexity and heterogeneity of the tumor-immune microenvironment (TIME). Strategies that simultaneously and synergistically engage multiple immune cells in TIME remain highly desirable but challenging. Herein, we report a multimodal and programmable platform that enables the integration of multiple therapeutic modules into single agents for tumor-targeted co-engagement of multiple immune cells within TIME. We developed the triple orthogonal linker (T-Linker) technology to integrate various therapeutic small molecules and biomolecules as multimodal targeting chimeras (Multi-TACs). The EGFR-CD3-PDL1 Multi-TAC facilitated T-dendritic cell co-engagement to target solid tumors with excellent efficacy, as demonstrated in vitro, in several humanized mouse models and in patient-derived tumor models. Furthermore, Multi-TACs were constructed to coordinate T cells with other immune cell types. The highly modular and programmable feature of our Multi-TACs may find broad applications in immunotherapy and beyond.
Collapse
Affiliation(s)
- Feng Lin
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Shenyi Yin
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Zijian Zhang
- National Resource Center for Mutant Mice, MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, Medical School of Nanjing University, Nanjing 210061, China
| | - Ying Yu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Haoming Fang
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Zhen Liang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Rujie Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Haitao Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Jianjie Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Medical Oncology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Kunxia Cao
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Weiming Guo
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Shan Qin
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Yuxuan Zhang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Chenghao Lu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Han Li
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Shibo Liu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Heng Zhang
- Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Buqing Ye
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Jian Lin
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China.
| | - Yan Li
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210061, China; National Resource Center for Mutant Mice, MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, Medical School of Nanjing University, Nanjing 210061, China.
| | - Xiaozheng Kang
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, China.
| | - Jianzhong Jeff Xi
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.
| | - Peng R Chen
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Shenzhen Bay Laboratory, Shenzhen 518055, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
11
|
Yang Y, Wu A, Deng AN, Liu H, Lan Q, Mazhar M, Xue JY, Chen MT, Luo G, Liu MN. Macrophages after myocardial infarction: Mechanisms for repairing and potential as therapeutic approaches. Int Immunopharmacol 2024; 143:113562. [PMID: 39536484 DOI: 10.1016/j.intimp.2024.113562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/20/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Macrophages - one of the crucial immune cells, are recruited to the cardiac tissue by chemokines, cytokines and upregulated endothelial adhesion molecules after myocardial infarction (MI). During the course of inflammation in the cardiac tissue, necrotic cells and matrix debris is phagocytosed by M1 macrophages. During the resolution phase of cardiac inflammation, M2 macrophages promote cardiac recovery. Suppression or over expression of both the M1 and M2 macrophage subtypes significantly affect the reparation of infarction. Stem cells therapy, cytokine regulation and immune cells therapy are considered as effective interventions to regulate the phenotypic transformation of cardiac macrophages after MI. Intervention with macrophages in the myocardium has shown unique advantages. In this review, the mechanisms and role of macrophages in the development of MI are elaborated in detail, the promising therapeutic methods for regulating macrophage phenotypes, their limitations and possible future research directions are discussed.
Collapse
Affiliation(s)
- You Yang
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Ai Wu
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - An-Ni Deng
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Hao Liu
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Qi Lan
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Maryam Mazhar
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Jin-Yi Xue
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Ming-Tai Chen
- Department of Cardiovascular Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China.
| | - Gang Luo
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| | - Meng-Nan Liu
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
12
|
Aquino CO, Pereira FM, Frota ACC, Hofer CB, Milagres LG, Manfro WFP. Innate-immune cell distribution in pediatric HIV patients and uninfected controls. Rev Inst Med Trop Sao Paulo 2024; 66:e75. [PMID: 39699513 PMCID: PMC11654119 DOI: 10.1590/s1678-9946202466075] [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: 06/25/2024] [Accepted: 11/07/2024] [Indexed: 12/20/2024] Open
Abstract
Innate immune cells are important players during an infection. The frequency of monocytes, myeloid-derived suppressor cells (MDSCs), natural killer (NK), and NKT cells were assessed in blood samples of children and adolescents living with HIV (CALHIV) and HIV-uninfected (HU) children. Blood samples from 10 CALHIV (treated or not) and six HU individuals were collected for approximately one year. Flow cytometry was employed to phenotypically characterize cell populations. We found a lower frequency of classical monocytes in CALHIV patients compared to the HU group (35.75% vs. 62.60%, respectively) but a higher frequency of CD56-CD16+ NK cells in CALHIV patients compared to the HU group (1.45% vs. 0.44%, respectively). At baseline, the frequency of monocytic-MDSCs inversely correlated with CD56dimCD16+ NK cells (r= -0.73, p=0.020), CD56-CD16+ NK cells (r= -0.78, p=0.010), and with intermediate monocytes (r= -0.71, p=0.027) in the CALHIV group. We also found a negative correlation between CD56++CD16+- and CD56dimCD16+ NK cells with CD4 T cells frequency at baseline. The results suggest an alteration in the innate compartment of the CALHIV cohort, which may contribute to their susceptibility to infections.
Collapse
Affiliation(s)
- Cynthia Oliveira Aquino
- Universidade do Estado do Rio de Janeiro, Departamento de Microbiologia, Imunologia e Parasitologia, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Mariz Pereira
- Universidade do Estado do Rio de Janeiro, Departamento de Microbiologia, Imunologia e Parasitologia, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Cristina Cisne Frota
- Universidade Federal do Rio de Janeiro, Instituto de Puericultura e Pediatria Martagão Gesteira, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cristina Barroso Hofer
- Universidade Federal do Rio de Janeiro, Instituto de Puericultura e Pediatria Martagão Gesteira, Rio de Janeiro, Rio de Janeiro, Brazil
- Universidade Federal do Rio de Janeiro, Departamento de Medicina Preventiva, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucimar Gonçalves Milagres
- Universidade do Estado do Rio de Janeiro, Departamento de Microbiologia, Imunologia e Parasitologia, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wânia Ferraz Pereira Manfro
- Universidade do Estado do Rio de Janeiro, Departamento de Microbiologia, Imunologia e Parasitologia, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Zheng WR, Dan JY, Huo N, Zhang Z, Hou LH. Characteristics of the early innate response induced by the aerosolized Ad5-vectored COVID-19 vaccine. MOLECULAR BIOMEDICINE 2024; 5:64. [PMID: 39633154 PMCID: PMC11618260 DOI: 10.1186/s43556-024-00232-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/31/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Affiliation(s)
- Wan-Ru Zheng
- School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Jun-Yan Dan
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Nan Huo
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Zhe Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Li-Hua Hou
- School of Medicine, Zhejiang University, Hangzhou, 310058, China.
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China.
| |
Collapse
|
14
|
Xiao X, Ding Z, Shi Y, Zhang Q. Causal Role of Immune Cells in Chronic Obstructive Pulmonary Disease: A Two-Sample Mendelian Randomization Study. COPD 2024; 21:2327352. [PMID: 38573027 DOI: 10.1080/15412555.2024.2327352] [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: 10/31/2023] [Accepted: 02/27/2024] [Indexed: 04/05/2024]
Abstract
Accumulating evidence has highlighted the importance of immune cells in the pathogenesis of chronic obstructive pulmonary disease (COPD). However, the understanding of the causal association between immunity and COPD remains incomplete due to the existence of confounding variables. In this study, we employed a two-sample Mendelian randomization (MR) analysis, utilizing the genome-wide association study database, to investigate the causal association between 731 immune-cell signatures and the susceptibility to COPD from a host genetics perspective. To validate the consistency of our findings, we utilized MR analysis results of lung function data to assess directional concordance. Furthermore, we employed MR-Egger intercept tests, Cochrane's Q test, MR-PRESSO global test, and "leave-one-out" sensitivity analyses to evaluate the presence of horizontal pleiotropy, heterogeneity, and stability, respectively. Inverse variance weighting results showed that seven immune phenotypes were associated with the risk of COPD. Analyses of heterogeneity and pleiotropy analysis confirmed the reliability of MR results. These results highlight the interactions between the immune system and the lungs. Further investigations into their mechanisms are necessary and will contribute to inform targeted prevention strategies for COPD.
Collapse
Affiliation(s)
- Xinru Xiao
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Ziqi Ding
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yujia Shi
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Qian Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| |
Collapse
|
15
|
Yu J, Shang C, Deng X, Jia J, Shang X, Wang Z, Zheng Y, Zhang R, Wang Y, Zhang H, Liu H, Liu WJ, Li H, Cao B. Time-resolved scRNA-seq reveals transcription dynamics of polarized macrophages with influenza A virus infection and antigen presentation to T cells. Emerg Microbes Infect 2024; 13:2387450. [PMID: 39129565 PMCID: PMC11370681 DOI: 10.1080/22221751.2024.2387450] [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: 03/25/2024] [Revised: 07/02/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
Throughout history, the influenza A virus has caused numerous devastating global pandemics. Macrophages, as pivotal innate immune cells, exhibit a wide range of immune functions characterized by distinct polarization states, reflecting their intricate heterogeneity. In this study, we employed the time-resolved single-cell sequencing technique coupled with metabolic RNA labelling to elucidate the dynamic transcriptional changes in distinct polarized states of bone marrow-derived macrophages (BMDMs) upon infection with the influenza A virus. Our approach not only captures the temporal dimension of transcriptional activity, which is lacking in conventional scRNA-seq methods, but also reveals that M2-polarized Arg1_macrophage cluster is the sole state supporting successful replication of influenza A virus. Furthermore, we identified distinct antigen presentation capabilities to CD4+ T and CD8+ T cells across diverse polarized states of macrophages. Notably, the M1 phenotype, exhibited by (BMDMs) and murine alveolar macrophages (AMs), demonstrated superior conventional and cross-presentation abilities for exogenous antigens, with a particular emphasis on cross-presentation capacity. Additionally, as CD8+ T cell differentiation progressed, M1 polarization exhibited an enhanced capacity for cross-presentation. All three phenotypes of BMDMs, including M1, demonstrated robust presentation to CD4+ regulatory T cells, while displaying limited ability to present to naive CD4+ T cells. These findings offer novel insights into the immunological regulatory mechanisms governing distinct polarized states of macrophages, particularly their roles in restricting the replication of influenza A virus and modulating antigen-specific T cell responses through innate immunity.
Collapse
Affiliation(s)
- Jiapei Yu
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Congcong Shang
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People’s Republic of China
| | - Xiaoyan Deng
- THU-PKU Joint Center for Life Sciences, Tsinghua University, Beijing, People’s Republic of China
| | - Ju Jia
- Department of Infectious Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xiao Shang
- THU-PKU Joint Center for Life Sciences, Tsinghua University, Beijing, People’s Republic of China
| | - Zeyi Wang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People’s Republic of China
| | - Ying Zheng
- Department of Pulmonary and Critical Care Medicine, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, People’s Republic of China
| | - Rongling Zhang
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People’s Republic of China
| | - Yeming Wang
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Hui Zhang
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, People’s Republic of China
| | - Hongyu Liu
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, People’s Republic of China
| | - William J. Liu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention,Beijing, People’s Republic of China
| | - Hui Li
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Bin Cao
- Department of Pulmonary and Critical Care Medicine, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing, People’s Republic of China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People’s Republic of China
- Department of Infectious Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
16
|
Xiong S, Li N, Shi S, Zhao Y, Chen J, Ruan M, Xu Y, Liu R, Wang S, Wang H. Structural characterization of a polysaccharide from Scutellaria baicalensis Georgi and its immune-enhancing properties on RAW264.7 cells. Int J Biol Macromol 2024; 283:137890. [PMID: 39571863 DOI: 10.1016/j.ijbiomac.2024.137890] [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: 07/30/2024] [Revised: 10/27/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
A novel polysaccharide SPS01-2 (87.5 kDa) was isolated from the roots of Scutellaria baicalensis Georgi. Monosaccharide composition revealed that SPS01-2 consists of rhamnose, arabinose, galactose, galacturonic acid, and glucuronic acid in ratio of 4.4: 67.1: 22.2: 6.3: 1.2. Further investigations using methylation, NMR, and mass spectrometry indicated that SPS01-2 is classified as a type II arabinogalactan (AG-II) with a minor presence of type I rhamnogalacturonan (RG-I). The core structure alternates between 1,2/1,2,4-α-L-Rhap and 1,4-α-D-GalpA, with branches including 1,3,6-β-D-Galp, 1,3-β-D-Galp, T-β-D-Galp, and T-α-L-Rhap. The RG-I regions are linked to 1,6-β-D-Galp, and 1,3,6-β-D-Galp units. Numerous arabinan branches, featuring multiple branching points, are attached to the O-3 position of galactose. Additionally, T-β-D-Galp, 1,6-β-D-Galp, and T-β-D-4-OMe-GlcpA are also linked to galactose in the backbone. Furthermore, SPS01-2 demonstrated potential immune-enhancing properties by dose-dependently increasing proliferation, phagocytosis, and the production of nitric oxide and cytokines (TNF-α, IL-6, and IL-1β) in RAW264.7 cells. It also enhanced the expression of CD80, CD86, and MHC-II at concentrations ranging from 5 to 200 μg/mL. Moreover, the immunostimulatory activity of SPS01-2 was significantly reduced when branch linkages were removed through partial acid hydrolysis. Our findings indicate that SPS01-2 could serve as a natural immunostimulant in the food and pharmaceutical sectors.
Collapse
Affiliation(s)
- Si Xiong
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Mate ria Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Ning Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China
| | - Songshan Shi
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Mate ria Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Yonglin Zhao
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Mate ria Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Jie Chen
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Mate ria Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Min Ruan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China
| | - Yongbin Xu
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Mate ria Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Ruimin Liu
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Mate ria Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Shunchun Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Mate ria Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China.
| | - Huijun Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Mate ria Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China.
| |
Collapse
|
17
|
Walther LM, Gideon A, Sauter C, Leist M, Wirtz PH. Peripheral Blood Leukocyte Subpopulation Changes in Reaction to an Acute Psychosocial Stressor as Compared to an Active Placebo-Stressor in Healthy Young Males: Mediating Effects of Major Stress-Reactive Endocrine Parameters. Cells 2024; 13:1941. [PMID: 39682690 DOI: 10.3390/cells13231941] [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: 09/01/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Psychosocial stress has been proposed to induce a redistribution of immune cells, but a comparison with an active placebo-psychosocial stress control condition is lacking so far. We investigated immune cell redistribution due to psychosocial stress compared to that resulting from an active placebo-psychosocial stress but otherwise identical control condition. Moreover, we tested for mediating effects of endocrine parameters and blood volume changes. The final study sample comprised 64 healthy young men who underwent either a psychosocial stress condition (Trier Social Stress Test; TSST; n = 38) or an active placebo-psychosocial stress control condition (PlacTSST; n = 26). Immune cell counts and hemoglobin, epinephrine, norepinephrine, ACTH, renin, and aldosterone levels, as well as those of saliva cortisol, were determined before and up to 30 min after the TSST/PlacTSST. The TSST induced greater increases in total leukocyte, monocyte, and lymphocyte levels as compared to the PlacTSST (p's ≤ 0.001), but in not granulocyte counts. Neutrophil granulocyte counts increased in reaction to both the TSST and PlacTSST (p's ≤ 0.001), while eosinophil and basophil granulocyte counts did not. The psychosocial stress-induced increases in immune cell counts from baseline to peak (i.e., +1 min after TSST cessation) were independently mediated by parallel increases in epinephrine (ab's ≤ -0.43; 95% CIs [LLs ≤ -0.66; ULs ≤ -0.09]). Subsequent decreases in immune cell counts from +1 min to +10 min after psychosocial stress cessation were mediated by parallel epinephrine, renin, and blood volume decreases (ab's ≥ 0.17; 95% CIs [LLs ≥ 0.02; ULs ≥ 0.35]). Our findings indicate that psychosocial stress specifically induces immune cell count increases in most leukocyte subpopulations that are not secondary to the physical or cognitive demands of the stress task. Increases in the number of circulating neutrophil granulocytes, however, are not psychosocial stress-specific and even occur in situations with a low probability of threat or harm. Our findings point to a major role of epinephrine in mediating stress-induced immune cell count increases and of epinephrine, renin, and blood volume changes in mediating subsequent immune cell count decreases from +1 min to +10 min after psychosocial stress cessation.
Collapse
Affiliation(s)
- Lisa-Marie Walther
- Biological Work and Health Psychology, University of Konstanz, 78457 Konstanz, Germany
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, 78457 Konstanz, Germany
| | - Angelina Gideon
- Biological Work and Health Psychology, University of Konstanz, 78457 Konstanz, Germany
| | - Christine Sauter
- Biological Work and Health Psychology, University of Konstanz, 78457 Konstanz, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Petra H Wirtz
- Biological Work and Health Psychology, University of Konstanz, 78457 Konstanz, Germany
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, 78457 Konstanz, Germany
| |
Collapse
|
18
|
Backer RA, Probst HC, Clausen BE. Multiparameter Flow Cytometric Analysis of the Conventional and Monocyte-Derived DC Compartment in the Murine Spleen. Vaccines (Basel) 2024; 12:1294. [PMID: 39591196 PMCID: PMC11598974 DOI: 10.3390/vaccines12111294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/04/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Dendritic cells (DCs) are present in almost all tissues, where they act as sentinels involved in innate recognition and the initiation of adaptive immune responses. The DC family consists of several cell lineages that are heterogenous in their development, phenotype, and function. Within these DC lineages, further subdivisions exist, resulting in smaller, less characterized subpopulations, each with its unique immunomodulatory capabilities. Given the interest in utilizing DC for experimental studies and for vaccination purposes, it becomes increasingly crucial to thoroughly classify and characterize these diverse DC subpopulations. This understanding is vital for comprehending their relative contribution to the initiation, regulation, and propagation of immune responses. To facilitate such investigation, we here provide an easy and ready-to-use multicolor flow cytometry staining panel for the analysis of conventional DC, plasmacytoid DC, and monocyte-derived DC populations isolated from mouse spleens. This adaptable panel can be easily customized for the analysis of other tissue-specific DC populations, providing a valuable tool for DC research.
Collapse
Affiliation(s)
- Ronald A. Backer
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Hans Christian Probst
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Institute for Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Björn E. Clausen
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| |
Collapse
|
19
|
Palacios-Berraquero ML, Rodriguez-Marquez P, Calleja-Cervantes ME, Berastegui N, Zabaleta A, Burgos L, Alignani D, San Martin-Uriz P, Vilas-Zornoza A, Rodriguez-Diaz S, Inoges S, Lopez-Diaz de Cerio A, Huerga S, Tamariz E, Rifon J, Alfonso-Pierola A, Lasarte JJ, Paiva B, Hernaez M, Rodriguez-Otero P, San-Miguel J, Ezponda T, Rodriguez-Madoz JR, Prosper F. Molecular mechanisms promoting long-term cytopenia after BCMA CAR-T therapy in multiple myeloma. Blood Adv 2024; 8:5479-5492. [PMID: 39058976 PMCID: PMC11532743 DOI: 10.1182/bloodadvances.2023012522] [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: 12/27/2023] [Revised: 06/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
ABSTRACT Hematologic toxicity is a common side effect of chimeric antigen receptor T-cell (CAR-T) therapies, being particularly severe among patients with relapsed or refractory multiple myeloma (MM). In this study, we characterized 48 patients treated with B-cell maturation antigen (BCMA) CAR-T cells to understand kinetics of cytopenia, identify predictive factors, and determine potential mechanisms underlying these toxicities. We observed that overall incidence of cytopenia was 95.7%, and grade >3 thrombocytopenia and neutropenia, 1 month after infusion, was observed in 57% and 53% of the patients, respectively, being still present after 1 year in 4 and 3 patients, respectively. Baseline cytopenia and high peak inflammatory markers were highly correlated with cytopenia that persisted up to 3 months. To determine potential mechanisms underlying cytopenias, we evaluated the paracrine effect of BCMA CAR-T cells on hematopoietic stem and progenitor cell (HSPC) differentiation using an ex vivo myeloid differentiation model. Phenotypic analysis showed that supernatants from activated CAR-T cells (spCAR) halted HSPC differentiation, promoting more immature phenotypes, which could be prevented with a combination of interferon γ, tumor necrosis factor α/β, transforming growth factor β, interleukin-6 (IL-6) and IL-17 inhibitors. Single-cell RNA sequencing demonstrated upregulation of transcription factors associated with early stages of hematopoietic differentiation in the presence of spCAR (GATA2, RUNX1, CEBPA) and a decrease in the activity of key regulons involved in neutrophil and monocytic maturation (ID2 and MAFB). These results suggest that CAR-T activation induces HSPC maturation arrest through paracrine effects and provides potential treatments to mitigate the severity of this toxicity.
Collapse
Affiliation(s)
- Maria Luisa Palacios-Berraquero
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Paula Rodriguez-Marquez
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Maria Erendira Calleja-Cervantes
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Nerea Berastegui
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Aintzane Zabaleta
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Leire Burgos
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Diego Alignani
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Patxi San Martin-Uriz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Amaia Vilas-Zornoza
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Saray Rodriguez-Diaz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Susana Inoges
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Immunology and Immunotherapy Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Ascensión Lopez-Diaz de Cerio
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Immunology and Immunotherapy Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Sofia Huerga
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Esteban Tamariz
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Jose Rifon
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Ana Alfonso-Pierola
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Juan Jose Lasarte
- Immunology and Immunotherapy Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Bruno Paiva
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Mikel Hernaez
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Data Science and Artificial Intelligence Institute, Universidad de Navarra, Pamplona, Spain
| | - Paula Rodriguez-Otero
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Jesus San-Miguel
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Teresa Ezponda
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Juan Roberto Rodriguez-Madoz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Felipe Prosper
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| |
Collapse
|
20
|
Pan L, Fu M, Tang XL, Ling Y, Su Y, Ge J. Kirenol Ameliorates Myocardial Ischemia-Reperfusion Injury by Promoting Mitochondrial Function and Inhibiting Inflammasome Activation. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07635-4. [PMID: 39531114 DOI: 10.1007/s10557-024-07635-4] [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] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Macrophage-mediated inflammation plays a crucial role in the pathophysiological process of myocardial ischemia/reperfusion (I/R) injury. Recent studies have highlighted the importance of mitochondrial function and inflammasome activation in the inflammatory process. Kirenol, a well-known natural compound, has been shown to regulate inflammation in various diseases. This study investigated whether Kirenol could exert anti-inflammatory effects on macrophages during myocardial I/R injury. METHODS Mouse myocardial I/R models were established by 45 min of ischemia followed by 24 h of reperfusion. Saline or Kirenol treatment was administered. In vivo assessments included the evaluation of cardiac function, infarcted area, and immune cell infiltration. Subsequently, bone marrow-derived macrophages (BMDMs) were isolated, and mitochondrial function and pyroptosis were assessed. Furthermore, the study compared the cardioprotective effects of Kirenol with a specific NOX1/NOX4 inhibitor, GKT137831. RESULTS Kirenol gavage improved cardiac function, decreased infarct area, and alleviated inflammatory infiltration in mice subjected to myocardial I/R injury. Mechanistically, Kirenol inhibited NOX1 and NOX4 and enhanced mitochondrial function, ultimately attenuating the pyroptosis of macrophages. The therapeutic effects of Kirenol and GKT137831 were not significantly different. CONCLUSION This study demonstrates that Kirenol mitigates myocardial I/R injury by inhibiting NOX1 and NOX4, restoring mitochondrial function, and ameliorating macrophage pyroptosis.
Collapse
Affiliation(s)
- Lei Pan
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Fenglin Road 180, Shanghai, 200032, Xuhui District, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Mingqiang Fu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Fenglin Road 180, Shanghai, 200032, Xuhui District, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiang-Lin Tang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Fenglin Road 180, Shanghai, 200032, Xuhui District, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Yunlong Ling
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Fenglin Road 180, Shanghai, 200032, Xuhui District, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Yangang Su
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Fenglin Road 180, Shanghai, 200032, Xuhui District, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
- NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Fenglin Road 180, Shanghai, 200032, Xuhui District, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Schwarz E, Benner B, Wesolowski R, Quiroga D, Good L, Sun SH, Savardekar H, Li J, Jung KJ, Duggan MC, Lapurga G, Shaffer J, Scarberry L, Konda B, Verschraegen C, Kendra K, Shah M, Rupert R, Monk P, Shah HA, Noonan AM, Bixel K, Hays J, Wei L, Pan X, Behbehani G, Hu Y, Elemento O, Chung D, Xin G, Blaser BW, Carson WE. Inhibition of Bruton's tyrosine kinase with PD-1 blockade modulates T cell activation in solid tumors. JCI Insight 2024; 9:e169927. [PMID: 39513363 PMCID: PMC11601564 DOI: 10.1172/jci.insight.169927] [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: 02/24/2023] [Accepted: 09/18/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUNDInhibition of Bruton's tyrosine kinase with ibrutinib blocks the function of myeloid-derived suppressor cells (MDSC). The combination of ibrutinib and nivolumab was tested in patients with metastatic solid tumors.METHODSSixteen patients received ibrutinib 420 mg p.o. daily with nivolumab 240 mg i.v. on days 1 and 15 of a 28-day cycle. The effect of ibrutinib and nivolumab on MDSC, the immune profile, and cytokine levels were measured. Single-cell RNA-Seq and T cell receptor sequencing of immune cells was performed.RESULTSCommon adverse events were fatigue and anorexia. Four patients had partial responses and 4 had stable disease at 3 months (average 6.5 months, range 3.5-14.6). Median overall survival (OS) was 10.8 months. Seven days of Bruton's tyrosine kinase (BTK) inhibition significantly increased the proportion of monocytic-MDSC (M-MDSC) and significantly decreased chemokines associated with MDSC recruitment and accumulation (CCL2, CCL3, CCL4, CCL13). Single-cell RNA-Seq revealed ibrutinib-induced downregulation of genes associated with MDSC-suppressive function (TIMP1, CXCL8, VEGFA, HIF1A), reduced MDSC interactions with exhausted CD8+ T cells, and decreased TCR repertoire diversity. The addition of nivolumab significantly increased circulating NK and CD8+ T cells and increased CD8+ T cell proliferation. Exploratory analyses suggest that MDSC and T cell gene expression and TCR repertoire diversity were differentially affected by BTK inhibition according to patient response.CONCLUSIONIbrutinib and nivolumab were well tolerated and affected MDSC and T cell function in patients with solid metastatic tumors.TRIAL REGISTRATIONClinicalTrials.gov NCT03525925.FUNDINGNIH; National Cancer Institute Cancer; National Center for Advancing Translational Sciences; Pelotonia.
Collapse
Affiliation(s)
| | | | - Robert Wesolowski
- Comprehensive Cancer Center
- Division of Medical Oncology, Department of Internal Medicine
| | - Dionisia Quiroga
- Comprehensive Cancer Center
- Division of Medical Oncology, Department of Internal Medicine
| | | | - Steven H. Sun
- Comprehensive Cancer Center
- Division of Surgical Oncology, Department of Surgery
| | | | - Jianying Li
- Comprehensive Cancer Center
- Department of Biomedical Informatics, College of Medicine
| | - Kyeong Joo Jung
- Comprehensive Cancer Center
- Department of Biomedical Informatics, College of Medicine
| | | | | | | | | | - Bhavana Konda
- Comprehensive Cancer Center
- Division of Medical Oncology, Department of Internal Medicine
| | - Claire Verschraegen
- Comprehensive Cancer Center
- Division of Medical Oncology, Department of Internal Medicine
| | - Kari Kendra
- Comprehensive Cancer Center
- Division of Medical Oncology, Department of Internal Medicine
| | - Manisha Shah
- Comprehensive Cancer Center
- Division of Medical Oncology, Department of Internal Medicine
| | - Robert Rupert
- Comprehensive Cancer Center
- Division of Medical Oncology, Department of Internal Medicine
| | - Paul Monk
- Comprehensive Cancer Center
- Division of Medical Oncology, Department of Internal Medicine
| | - Hiral A. Shah
- Comprehensive Cancer Center
- Division of Medical Oncology, Department of Internal Medicine
| | - Anne M. Noonan
- Comprehensive Cancer Center
- Division of Medical Oncology, Department of Internal Medicine
| | - Kristin Bixel
- Comprehensive Cancer Center
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology
| | - John Hays
- Comprehensive Cancer Center
- Division of Medical Oncology, Department of Internal Medicine
| | - Lai Wei
- Comprehensive Cancer Center
- Department of Biomedical Informatics, College of Medicine
| | | | - Gregory Behbehani
- Comprehensive Cancer Center
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Yang Hu
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA
| | - Dongjun Chung
- Comprehensive Cancer Center
- Department of Biomedical Informatics, College of Medicine
| | - Gang Xin
- Comprehensive Cancer Center
- Department of Biomedical Informatics, College of Medicine
| | - Bradley W. Blaser
- Comprehensive Cancer Center
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - William E. Carson
- Comprehensive Cancer Center
- Division of Surgical Oncology, Department of Surgery
| |
Collapse
|
22
|
Chen T, Yang Y. Immunologic and inflammatory pathogenesis of chronic coronary syndromes: A review. Medicine (Baltimore) 2024; 103:e40354. [PMID: 39496055 PMCID: PMC11537619 DOI: 10.1097/md.0000000000040354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/15/2024] [Indexed: 11/06/2024] Open
Abstract
Chronic coronary syndrome (CCS) is a major cause of progression to acute coronary syndrome. Due to its insidious onset and complex etiology, this condition is often underestimated and insufficiently recognized, and traditional interventions for risk factors do not effectively control the disease progression. Current research suggests that immune and inflammatory pathways contribute to atherosclerosis and its clinical complications, thereby triggering the progression of CCS to acute coronary syndrome. This article primarily reviews the possible mechanisms of immune and inflammatory responses in CCS, with the aim of providing references for the diagnosis, treatment, and prevention of CCS.
Collapse
Affiliation(s)
- Tingting Chen
- Dali University School of Clinical Medicine, Yunnan, China
| | - Ying Yang
- Department of Cardiology, The First Affiliated Hospital of Dali University, Yunnan, China
| |
Collapse
|
23
|
Wang S, Zhang Z, Wang J, Lou Y, Zhu Y, You J, Liu P, Xu LX. Neutrophils promote the activation of monocytes via ROS to boost systemic antitumor immunity after cryo-thermal therapy. Front Immunol 2024; 15:1445513. [PMID: 39555061 PMCID: PMC11563809 DOI: 10.3389/fimmu.2024.1445513] [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: 06/07/2024] [Accepted: 10/14/2024] [Indexed: 11/19/2024] Open
Abstract
Background The characteristics of the tumor immunosuppressive microenvironment represent a major challenge that limits the efficacy of immunotherapy. Our previous results suggested that cryo-thermal therapy, a tumor ablation system developed in our laboratory, promotes macrophage M1-type polarization and the complete maturation of DCs to remodel the immunosuppressive environment. However, the cells that respond promptly to CTT have not yet been identified. CTT can cause extensive cell death and the release of danger-associated molecular patterns and antigens. Neutrophils are the first white blood cells recruited to sites of damage and acute inflammation. Therefore, we hypothesized that neutrophils are the initial cells that respond to CTT and are involved in the subsequent establishment of antitumor immunity. Methods In this study, we examined the kinetics of neutrophil recruitment after CTT via flow cytometry and immunofluorescence staining and explored the effect of neutrophils on the establishment of systemic antitumor immunity by in vivo neutrophil depletion and in vitro co-culture assays. Results We found that CTT led to a rapid and strong proinflammatory neutrophil response, which was essential for the long-term survival of mice. CTT-induced neutrophils promoted the activation of monocytes via reactive oxygen species and further upregulated the expression of IFN-γ and cytotoxic molecules in T and NK cells. Adoptive neutrophil transfer further enhanced the antitumor efficacy of CTT in tumor models of spontaneous and experimental metastasis. Conclusion These results reveal the important role of neutrophil‒monocyte interactions in the development of anti-tumor immunity and highlight that CTT could be used as an immunotherapy for targeting neutrophils and monocytes to enhance antitumor immunity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ping Liu
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Lisa X. Xu
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
24
|
Sabatini A, Lucidi M, Ciolfi S, Vuotto C, De Bardi M, Visca P, Battistini L, Visaggio D, Volpe E. Innate immune mechanisms promote human response to Acinetobacter baumannii infection. Eur J Immunol 2024; 54:e2451170. [PMID: 39072714 DOI: 10.1002/eji.202451170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
Acinetobacter baumannii is an opportunistic Gram-negative bacterium representing one of the leading causes of ventilator-associated pneumonia. The development of pneumonia results from a complex interplay between pathogens and pulmonary innate mucosal immunity. Therefore, the knowledge of the host immune responses is pivotal for the development of effective therapeutics to treat A. baumannii infections. Previous studies were conducted using cell lines and animal models, but a comprehensive understanding of the interaction between A. baumannii and primary human immune cells is still lacking. To bridge this gap, we investigated the response of primary monocytes, macrophages, and dendritic cells to the A. baumannii-type strain and an epidemic clinical isolate. We found that all immune cells trigger different responses when interacting with A. baumannii. In particular, macrophages and monocytes mediate bacterial clearance, whereas monocytes and dendritic cells activate a late response through the production of cytokines, chemokines, and the expression of co-stimulatory molecules. The epidemic strain induces lower expression of interleukin-10 and CD80 compared with the type strain, potentially constituting two immune evasion strategies.
Collapse
Affiliation(s)
- Andrea Sabatini
- Molecular Neuroimmunology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Massimiliano Lucidi
- Department of Science, University Roma Tre, Rome, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Serena Ciolfi
- Molecular Neuroimmunology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Claudia Vuotto
- Neuromicrobiology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Marco De Bardi
- Neuroimmunology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Paolo Visca
- Department of Science, University Roma Tre, Rome, Italy
- Molecular Microbiology and Microbiomics, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Luca Battistini
- Neuroimmunology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Daniela Visaggio
- Department of Science, University Roma Tre, Rome, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
- Molecular Microbiology and Microbiomics, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Elisabetta Volpe
- Molecular Neuroimmunology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
25
|
Dyba M, Berezenko V, Zabara D, Bezpala A, Donskoi B. Monocyte subpopulations in children with autoimmune liver disease. Pathol Res Pract 2024; 263:155622. [PMID: 39357182 DOI: 10.1016/j.prp.2024.155622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Patients with autoimmune liver diseases require individualized long-term immunosuppressive therapy, whose discontinuation is possible after complete histological remission and that requires repeated liver biopsy. In view of this, the search for non-invasive markers is essential for patients with autoimmune liver disease. PURPOSE The purpose of this research is to assess the possibility of predicting the recurrence of autoimmune liver disease in children. METHOD The biological material used in the study was blood serum from 80 children diagnosed with autoimmune hepatitis and autoimmune sclerosing cholangitis. Patients were divided into four groups according to disease activity and therapeutic approach. RESULTS The percentage of monocyte subpopulations was determined by flow cytometry, and disease activity, inflammation, and fibrosis markers were analyzed to study the relationship and diagnostic value of the parameters studied in detail. The results of the study indicate a significant relationship between disease activity and changes in the distribution of the percentage of monocyte subpopulations in the blood. The percentage of intermediate CD14++/CD16+ monocytes was found to correlate with disease activity, and non-classical CD14lowCD16+ monocytes were found to be of high diagnostic value in the diagnosis of disease relapse. CONCLUSIONS These findings not only expand the understanding of the pathogenesis of autoimmune liver disease but also point to the prospects of using monocyte subpopulations as potential biomarkers for predicting relapse, contributing to the development of more effective clinical management strategies.
Collapse
Affiliation(s)
- Maryna Dyba
- Department of Hepatology and Comorbidities in Children, Institute of Pediatrics, Obstetrics and Gynecology of the National Academy of Medical Sciences of Ukraine, Kyiv 04050, Ukraine.
| | - Valentyna Berezenko
- Department of Hepatology and Comorbidities in Children, Institute of Pediatrics, Obstetrics and Gynecology of the National Academy of Medical Sciences of Ukraine, Kyiv 04050, Ukraine
| | - Dariia Zabara
- Laboratory of Immunology, Institute of Pediatrics, Obstetrics and Gynecology of the National Academy of Medical Sciences of Ukraine, Kyiv 04050, Ukraine
| | - Anna Bezpala
- Department of Hepatology and Comorbidities in Children, Institute of Pediatrics, Obstetrics and Gynecology of the National Academy of Medical Sciences of Ukraine, Kyiv 04050, Ukraine
| | - Boris Donskoi
- Laboratory of Immunology, Institute of Pediatrics, Obstetrics and Gynecology of the National Academy of Medical Sciences of Ukraine, Kyiv 04050, Ukraine
| |
Collapse
|
26
|
Häberle J, Siri B, Dionisi‐Vici C. Quo vadis ureagenesis disorders? A journey from 90 years ago into the future. J Inherit Metab Dis 2024; 47:1120-1128. [PMID: 38837457 PMCID: PMC11586591 DOI: 10.1002/jimd.12763] [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: 02/05/2024] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 06/07/2024]
Abstract
The pathway of ammonia disposal in the mammalian organism has been described in 1932 as a metabolic cycle present in the liver in different compartments. In 1958, the first human disorder affecting this pathway was described as a genetic condition leading to cognitive impairment and constant abnormalities of amino acid metabolism. Since then, defects in all enzymes and transporters of the urea cycle have been described, referring to them as primary urea cycle disorders causing primary hyperammonemia. In addition, there is a still increasing list of conditions that impact on the function of the urea cycle by various mechanisms, hereby leading to secondary hyperammonemia. Despite great advances in understanding the molecular background and the biochemical specificities of both primary and secondary hyperammonemias, there remain many open questions: we do not fully understand the pathophysiology in many of the conditions; we do not always understand the highly variable clinical course of affected patients; we clearly appreciate the need for novel and improved diagnostic and therapeutic approaches. This study does look back to the beginning of the urea cycle (hi)story, briefly describes the journey through past decades, hereby illustrating advancements and knowledge gaps, and gives examples for the extremely broad perspective imminent to some of the defects of ureagenesis and allied conditions.
Collapse
Affiliation(s)
- Johannes Häberle
- Division of Metabolism and Children's Research CenterUniversity Children's Hospital Zurich, University of ZurichZurichSwitzerland
| | - Barbara Siri
- Division of Metabolic Diseases and HepatologyBambino Gesù Children's Hospital IRCCSRomeItaly
| | - Carlo Dionisi‐Vici
- Division of Metabolic Diseases and HepatologyBambino Gesù Children's Hospital IRCCSRomeItaly
| |
Collapse
|
27
|
Jernigan JE, Staley HA, Baty Z, Bolen ML, Gomes BN, Holt J, Cole CL, Neighbarger NK, Dheeravath K, Merchak AR, Menees KB, Coombes SA, Tansey MG. RGS10 Attenuates Systemic Immune Dysregulation Induced by Chronic Inflammatory Stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.620078. [PMID: 39554164 PMCID: PMC11566001 DOI: 10.1101/2024.10.24.620078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Regulator of G-protein signaling 10 (RGS10), a key homeostatic regulator of immune cells, has been implicated in multiple diseases associated with aging and chronic inflammation including Parkinson's Disease (PD). Interestingly, subjects with idiopathic PD display reduced levels of RGS10 in subsets of peripheral immune cells. Additionally, individuals with PD have been shown to have increased activated peripheral immune cells in cerebral spinal fluid (CSF) compared to age-matched healthy controls. However, it is unknown whether CSF-resident peripheral immune cells in individuals with PD also exhibit decreased levels of RGS10. Therefore, we performed an analysis of RGS10 levels in the proteomic database of the CSF from the Michael J. Fox Foundation Parkinson's Progression Markers Initiative (PPMI) study. We found that RGS10 levels are decreased in the CSF of individuals with PD compared to healthy controls and prodromal individuals. Moreover, we find that RGS10 levels decrease with age but not PD progression and that males have less RGS10 than females in PD. Importantly, studies have established an association between chronic systemic inflammation (CSI) and neurodegenerative diseases, such as PD, and known sources of CSI have been identified as risk factors for developing PD; however, the role of peripheral immune cell dysregulation in this process has been underexplored. As RGS10 levels are decreased in the CSF and circulating peripheral immune cells of individuals with PD, we hypothesized that RGS10 regulates peripheral immune cell responses to CSI prior to the onset of neurodegeneration. To test this, we induced CSI for 6 weeks in C57BL6/J mice and RGS10 KO mice to assess circulating and CNS-associated peripheral immune cell responses. We found that RGS10 deficiency synergizes with CSI to induce a bias for inflammatory and cytotoxic cell populations, a reduction in antigen presentation in peripheral blood immune cells, as well as in and around the brain that is most notable in males. These results highlight RGS10 as an important regulator of the systemic immune response to CSI and implicate RGS10 as a potential contributor to the development of immune dysregulation in PD.
Collapse
Affiliation(s)
- Janna E. Jernigan
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Hannah A. Staley
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Zachary Baty
- Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL USA
| | - MacKenzie L. Bolen
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Beatriz Nuñes Gomes
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Jenny Holt
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Cassandra L. Cole
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Noelle K. Neighbarger
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Kruthika Dheeravath
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Andrea R. Merchak
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Kelly B. Menees
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Stephen A. Coombes
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL USA
| | - Malú Gámez Tansey
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| |
Collapse
|
28
|
Zhang W, Zhang X, Qiu C, Zhang Z, Su KJ, Luo Z, Liu M, Zhao B, Wu L, Tian Q, Shen H, Wu C, Deng HW. An atlas of genetic effects on the monocyte methylome across European and African populations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.12.24311885. [PMID: 39211851 PMCID: PMC11361221 DOI: 10.1101/2024.08.12.24311885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Elucidating the genetic architecture of DNA methylation is crucial for decoding complex disease etiology. However, current epigenomic studies are often limited by incomplete methylation coverage and heterogeneous tissue samples. Here, we present the first comprehensive, multi-ancestry human methylome atlas of purified human monocytes, generated through integrated whole-genome bisulfite sequencing and whole-genome sequencing from 298 European Americans (EA) and 160 African Americans (AA). By analyzing over 25 million methylation sites, we identified 1,383,250 and 1,721,167 methylation quantitative trait loci (meQTLs) in cis- regions for EA and AA populations, respectively, revealing both shared (880,108 sites) and population-specific regulatory patterns. Furthermore, we developed population-specific DNAm imputation models, enabling methylome-wide association studies (MWAS) for 1,976,046 and 2,657,581 methylation sites in EA and AA, respectively. These models were validated through multi-ancestry analysis of 41 complex traits from the Million Veteran Program. The identified meQTLs, MWAS models, and data resources are freely available at www.gcbhub.org and https://osf.io/gct57/ .
Collapse
|
29
|
Gallerand A, Dolfi B, Stunault MI, Caillot Z, Castiglione A, Strazzulla A, Chen C, Heo GS, Luehmann H, Batoul F, Vaillant N, Dumont A, Pilot T, Merlin J, Zair FN, Gilleron J, Bertola A, Carmeliet P, Williams JW, Arguello RJ, Masson D, Dombrowicz D, Yvan-Charvet L, Doyen D, Haschemi A, Liu Y, Guinamard RR, Ivanov S. Glucose metabolism controls monocyte homeostasis and migration but has no impact on atherosclerosis development in mice. Nat Commun 2024; 15:9027. [PMID: 39424804 PMCID: PMC11489573 DOI: 10.1038/s41467-024-53267-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
Monocytes directly contribute to atherosclerosis development by their recruitment to plaques in which they differentiate into macrophages. In the present study, we ask how modulating monocyte glucose metabolism could affect their homeostasis and their impact on atherosclerosis. Here we investigate how circulating metabolites control monocyte behavior in blood, bone marrow and peripheral tissues of mice. We find that serum glucose concentrations correlate with monocyte numbers. In diet-restricted mice, monocytes fail to metabolically reprogram from glycolysis to fatty acid oxidation, leading to reduced monocyte numbers in the blood. Mechanistically, Glut1-dependent glucose metabolism helps maintain CD115 membrane expression on monocytes and their progenitors, and regulates monocyte migratory capacity by modulating CCR2 expression. Results from genetic models and pharmacological inhibitors further depict the relative contribution of different metabolic pathways to the regulation of CD115 and CCR2 expression. Meanwhile, Glut1 inhibition does not impact atherosclerotic plaque development in mouse models despite dramatically reducing blood monocyte numbers, potentially due to the remaining monocytes having increased migratory capacity. Together, these data emphasize the role of glucose uptake and intracellular glucose metabolism in controlling monocyte homeostasis and functions.
Collapse
Affiliation(s)
- Alexandre Gallerand
- Université Côte d'Azur, CNRS, LP2M, Nice, France.
- Université Côte d'Azur, INSERM, C3M, Nice, France.
| | - Bastien Dolfi
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | | | - Alexia Castiglione
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | - Chuqiao Chen
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Gyu Seong Heo
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Flora Batoul
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | | | - Thomas Pilot
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France
| | | | | | | | | | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute (LKI), KU Leuven, Leuven, 3000, Belgium
| | - Jesse W Williams
- Center for Immunology, Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Rafael J Arguello
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - David Masson
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France
| | - David Dombrowicz
- Univ.Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | | | - Denis Doyen
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Médecine Intensive Réanimation, Hôpital Pasteur, CHU de Nice, Nice, France
| | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rodolphe R Guinamard
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | - Stoyan Ivanov
- Université Côte d'Azur, CNRS, LP2M, Nice, France.
- Université Côte d'Azur, INSERM, C3M, Nice, France.
| |
Collapse
|
30
|
Najibi AJ, Lane RS, Sobral MC, Bovone G, Kang S, Freedman BR, Gutierrez Estupinan J, Elosegui-Artola A, Tringides CM, Dellacherie MO, Williams K, Ijaz H, Müller S, Turley SJ, Mooney DJ. Durable lymph-node expansion is associated with the efficacy of therapeutic vaccination. Nat Biomed Eng 2024; 8:1226-1242. [PMID: 38710838 PMCID: PMC11485260 DOI: 10.1038/s41551-024-01209-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/30/2024] [Indexed: 05/08/2024]
Abstract
Following immunization, lymph nodes dynamically expand and contract. The mechanical and cellular changes enabling the early-stage expansion of lymph nodes have been characterized, yet the durability of such responses and their implications for adaptive immunity and vaccine efficacy are unknown. Here, by leveraging high-frequency ultrasound imaging of the lymph nodes of mice, we report more potent and persistent lymph-node expansion for animals immunized with a mesoporous silica vaccine incorporating a model antigen than for animals given bolus immunization or standard vaccine formulations such as alum, and that durable and robust lymph-node expansion was associated with vaccine efficacy and adaptive immunity for 100 days post-vaccination in a mouse model of melanoma. Immunization altered the mechanical and extracellular-matrix properties of the lymph nodes, drove antigen-dependent proliferation of immune and stromal cells, and altered the transcriptional features of dendritic cells and inflammatory monocytes. Strategies that robustly maintain lymph-node expansion may result in enhanced vaccination outcomes.
Collapse
Affiliation(s)
- Alexander J Najibi
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Ryan S Lane
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Miguel C Sobral
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Giovanni Bovone
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Shawn Kang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Benjamin R Freedman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Joel Gutierrez Estupinan
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Alberto Elosegui-Artola
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
- Institute for Bioengineering of Catalonia, Barcelona, Spain
| | - Christina M Tringides
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
- Harvard Program in Biophysics, Harvard University, Cambridge, MA, USA
| | - Maxence O Dellacherie
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Katherine Williams
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Hamza Ijaz
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Sören Müller
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Shannon J Turley
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.
| |
Collapse
|
31
|
Hu Y, Tang J, Xu Q, Fang Z, Li R, Yang M, Zhao J, Chen X. Role of pyruvate kinase M2 in regulating sepsis (Review). Mol Med Rep 2024; 30:185. [PMID: 39155878 DOI: 10.3892/mmr.2024.13309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/29/2024] [Indexed: 08/20/2024] Open
Abstract
Glycolysis occurs in all living organisms as a form of energy supply. Pyruvate kinase M2 (PKM2) is one of the rate‑limiting enzymes in the glycolytic process. PKM2 is considered to serve an important role in several terminal diseases, including sepsis. However, to the best of our knowledge, the specific mechanistic role of PKM2 in sepsis remains to be systematically summarised. Therefore, the present review aims to summarise the roles of PKM2 in sepsis progression. In addition, potential treatment strategies for patients with sepsis are discussed. The present review hopes to lay the groundwork for studying the role of PKM2 and developing therapeutic strategies against metabolic disorders that occur during sepsis.
Collapse
Affiliation(s)
- Yifei Hu
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Jing Tang
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Qiao Xu
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Zenghui Fang
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Rongqing Li
- Department of Clinical Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu 225000, P.R. China
| | - Mengxuan Yang
- Department of Clinical Laboratory, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Jie Zhao
- Department of Clinical Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu 225000, P.R. China
| | - Xin Chen
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| |
Collapse
|
32
|
Pries R, Plötze-Martin K, Lange C, Behn N, Werner L, Bruchhage KL, Steffen A. Improved levels of checkpoint molecule PD-L1 on peripheral blood monocyte subsets in obstructive sleep apnea syndrome patients upon hypoglossal nerve stimulation. J Sleep Res 2024; 33:e14178. [PMID: 38385644 DOI: 10.1111/jsr.14178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/02/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024]
Abstract
Oxidative stress in patients suffering from obstructive sleep apnea syndrome (OSAS) is associated with a low-grade systemic inflammation, immune disturbance, and increased invasion of monocytes into the endothelium. Besides continuous positive airway pressure (PAP), hypoglossal nerve stimulation (HNS) has become a promising treatment option for patients with OSAS. We aimed to analyse the influence of HNS therapy on the cellular characteristics relevant for adhesion and immune regulation of circulating CD14/CD16 monocyte subsets. Whole blood flow cytometric measurements were performed to analyse the expression levels of different adhesion molecules and checkpoint molecule PD-L1 (programmed death-ligand 1) in connection with pro-inflammatory plasma cytokine IL-8 and the clinical values of BMI (body mass index), AHI (apnea-hypopnea index), ODI (oxygen desaturation index), and ESS (Epworth sleepiness scale) upon HNS treatment. Hypoglossal nerve stimulation treatment significantly improved the expression of adhesion molecule CD162 (P-selectin receptor) on non-classical monocytes and significantly downregulated the expression of PD-L1 on all three monocyte subsets. We conclude that the holistic improvement of different parameters such as the oxygenation of the peripheral blood, a reduced systemic inflammation, and the individual sleeping situation upon HNS respiratory support, leads to an improved immunologic situation.
Collapse
Affiliation(s)
- Ralph Pries
- Department of Otorhinolaryngology, University of Luebeck, Luebeck, Germany
| | | | - Christian Lange
- Department of Otorhinolaryngology, University of Luebeck, Luebeck, Germany
| | - Nicole Behn
- Department of Otorhinolaryngology, University of Luebeck, Luebeck, Germany
| | - Lotte Werner
- Department of Otorhinolaryngology, University of Luebeck, Luebeck, Germany
| | | | - Armin Steffen
- Department of Otorhinolaryngology, University of Luebeck, Luebeck, Germany
| |
Collapse
|
33
|
Su C, Lee D, Jin P, Zhang J. Cell-type-specific mapping of enhancers and target genes from single-cell multimodal data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614814. [PMID: 39386519 PMCID: PMC11463474 DOI: 10.1101/2024.09.24.614814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Mapping enhancers and target genes in disease-related cell types has provided critical insights into the functional mechanisms of genetic variants identified by genome-wide association studies (GWAS). However, most existing analyses rely on bulk data or cultured cell lines, which may fail to identify cell-type-specific enhancers and target genes. Recently, single-cell multimodal data measuring both gene expression and chromatin accessibility within the same cells have enabled the inference of enhancer-gene pairs in a cell-type-specific and context-specific manner. However, this task is challenged by the data's high sparsity, sequencing depth variation, and the computational burden of analyzing a large number of enhancer-gene pairs. To address these challenges, we propose scMultiMap, a statistical method that infers enhancer-gene association from sparse multimodal counts using a joint latent-variable model. It adjusts for technical confounding, permits fast moment-based estimation and provides analytically derived p -values. In systematic analyses of blood and brain data, scMultiMap shows appropriate type I error control, high statistical power with greater reproducibility across independent datasets and stronger consistency with orthogonal data modalities. Meanwhile, its computational cost is less than 1% of existing methods. When applied to single-cell multimodal data from postmortem brain samples from Alzheimer's disease (AD) patients and controls, scMultiMap gave the highest heritability enrichment in microglia and revealed new insights into the regulatory mechanisms of AD GWAS variants in microglia.
Collapse
Affiliation(s)
- Chang Su
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Dongsoo Lee
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Peng Jin
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Jingfei Zhang
- Information Systems and Operations Management, Emory University, Atlanta, GA, USA
| |
Collapse
|
34
|
Kang N, Kim J, Kwon M, Son Y, Eo SK, Baryawno N, Kim BS, Yoon S, Oh SO, Lee D, Kim K. Blockade of mTORC1 via Rapamycin Suppresses 27-Hydroxycholestrol-Induced Inflammatory Responses. Int J Mol Sci 2024; 25:10381. [PMID: 39408711 PMCID: PMC11477202 DOI: 10.3390/ijms251910381] [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: 08/21/2024] [Revised: 09/15/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Atherosclerosis is characterized by the deposition and accumulation of extracellular cholesterol and inflammatory cells in the arterial blood vessel walls, and 27-hydroxycholesterol (27OHChol) is the most abundant cholesterol metabolite. 27OHChol is an oxysterol that induces immune responses, including immune cell activation and chemokine secretion, although the underlying mechanisms are not fully understood. In this study, we investigated the roles of the mechanistic target of rapamycin (mTOR) in 27HChol-induced inflammation using rapamycin. Treating monocytic cells with rapamycin effectively reduced the expression of CCL2 and CD14, which was involved with the increased immune response by 27OHChol. Rapamycin also suppressed the phosphorylation of S6 and 4EBP1, which are downstream of mTORC1. Additionally, it also alleviates the increase in differentiation markers into macrophage. These results suggest that 27OHChol induces inflammation by activating the mTORC1 signaling pathway, and rapamycin may be useful for the treatment of atherosclerosis-related inflammation involving 27OHchol.
Collapse
Affiliation(s)
- Nakyung Kang
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| | - Jaesung Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| | - Munju Kwon
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Yonghae Son
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| | - Seong-Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea;
| | - Ninib Baryawno
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institute, 17177 Stockholm, Sweden;
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| |
Collapse
|
35
|
Li J, Yang G, Liu J, Li G, Zhou H, He Y, Fei X, Zhao D. Integrating transcriptomics, eQTL, and Mendelian randomization to dissect monocyte roles in severe COVID-19 and gout flare. Front Genet 2024; 15:1385316. [PMID: 39385934 PMCID: PMC11461236 DOI: 10.3389/fgene.2024.1385316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction There are considerable similarities between the pathophysiology of gout flare and the dysregulated inflammatory response in severe COVID-19 infection. Monocytes are the key immune cells involved in the pathogenesis of both diseases. Therefore, it is critical to elucidate the molecular basis of the function of monocytes in gout and COVID-19 in order to develop more effective therapeutic approaches. Methods The single-cell RNA sequencing (scRNA-seq), large-scale genome-wide association studies (GWAS), and expression quantitative trait loci (eQTL) data of gout and severe COVID-19 were comprehensively analyzed. Cellular heterogeneity and intercellular communication were identified using the scRNA-seq datasets, and the monocyte-specific differentially expressed genes (DEGs) between COVID-19, gout and normal subjects were screened. In addition, the correlation of the DEGs with severe COVID-19 and gout flare was analyzed through GWAS statistics and eQTL data. Results The scRNA-seq analysis exhibited that the proportion of classical monocytes was increased in both severe COVID-19 and gout patient groups compared to healthy controls. Differential expression analysis and MR analysis showed that NLRP3 was positively associated with the risk of severe COVID-19 and involved 11 SNPs, of which rs4925547 was not significantly co-localized. In contrast, IER3 was positively associated with the risk of gout and involved 9 SNPs, of which rs1264372 was significantly co-localized. Discussion Monocytes have a complex role in gout flare and severe COVID-19, which underscores the potential mechanisms and clinical significance of the interaction between the two diseases.
Collapse
Affiliation(s)
- Jiajia Li
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Guixian Yang
- Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Junnan Liu
- Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Guofeng Li
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Huiling Zhou
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yuan He
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xinru Fei
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Dongkai Zhao
- Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
36
|
Jin Y, Wu Y, Reuben A, Zhu L, Gay CM, Wu Q, Zhou X, Mo H, Zheng Q, Ren J, Fang Z, Peng T, Wang N, Ma L, Fan Y, Song H, Zhang J, Chen M. Single-cell and spatial proteo-transcriptomic profiling reveals immune infiltration heterogeneity associated with neuroendocrine features in small cell lung cancer. Cell Discov 2024; 10:93. [PMID: 39231924 PMCID: PMC11375181 DOI: 10.1038/s41421-024-00703-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 06/23/2024] [Indexed: 09/06/2024] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive pulmonary neuroendocrine malignancy featured by cold tumor immune microenvironment (TIME), limited benefit from immunotherapy, and poor survival. The spatial heterogeneity of TIME significantly associated with anti-tumor immunity has not been systemically studied in SCLC. We performed ultra-high-plex Digital Spatial Profiling on 132 tissue microarray cores from 44 treatment-naive limited-stage SCLC tumors. Incorporating single-cell RNA-sequencing data from a local cohort and published SCLC data, we established a spatial proteo-transcriptomic landscape covering over 18,000 genes and 60 key immuno-oncology proteins that participate in signaling pathways affecting tumorigenesis, immune regulation, and cancer metabolism across 3 pathologically defined spatial compartments (pan-CK-positive tumor nest; CD45/CD3-positive tumor stroma; para-tumor). Our study depicted the spatial transcriptomic and proteomic TIME architecture of SCLC, indicating clear intra-tumor heterogeneity dictated via canonical neuroendocrine subtyping markers; revealed the enrichment of innate immune cells and functionally impaired B cells in tumor nest and suggested potentially important immunoregulatory roles of monocytes/macrophages. We identified RE1 silencing factor (REST) as a potential biomarker for SCLC associated with low neuroendocrine features, more active anti-tumor immunity, and prolonged survival.
Collapse
Affiliation(s)
- Ying Jin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, China
| | - Yuefeng Wu
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE), School of Medicine, Zhejiang University, Haining, Zhejiang, China
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Liang Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Carl M Gay
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qingzhe Wu
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xintong Zhou
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haomin Mo
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qi Zheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junyu Ren
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhaoyuan Fang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE), School of Medicine, Zhejiang University, Haining, Zhejiang, China
| | - Teng Peng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Nan Wang
- Cosmos Wisdom Biotech Co. Ltd., Hangzhou, Zhejiang, China
| | - Liang Ma
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yun Fan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Hai Song
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China.
- Center for Oncology Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, China.
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Ming Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co., Ltd, Guangzhou, Guangdong, China.
| |
Collapse
|
37
|
Minote M, Sato W, Kimura K, Kimura A, Lin Y, Okamoto T, Takahashi R, Yamamura T. High frequency of circulating non-classical monocytes is associated with stable remission in relapsing-remitting multiple sclerosis. Immunol Med 2024; 47:151-165. [PMID: 38539051 PMCID: PMC11346389 DOI: 10.1080/25785826.2024.2331271] [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: 01/29/2024] [Accepted: 03/07/2024] [Indexed: 08/23/2024] Open
Abstract
'No evidence of disease activity (NEDA)', judged by clinical and radiological findings, is a therapeutic goal in patients with multiple sclerosis (MS). It is, however, unclear if distinct biological mechanisms contribute to the maintenance of NEDA. To clarify the immunological background of long-term disease stability defined by NEDA, circulating immune cell subsets in patients with relapsing-remitting MS (RRMS) were analyzed using flow cytometry. Patients showing long-term NEDA (n = 31) had significantly higher frequencies of non-classical monocytes (NCMs) (6.1% vs 1.4%) and activated regulatory T cells (Tregs; 2.1% vs 1.6%) than those with evidence of disease activity (n = 8). The NCM frequency and NCMs to classical monocytes ratio (NCM/CM) positively correlated with activated Treg frequency and duration of NEDA. Co-culture assays demonstrated that NCMs could increase the frequency of activated Tregs and the expression of PD-L1, contributing to development of Tregs, was particularly high in NCMs from patients with NEDA. Collectively, NCMs contribute to stable remission in patients with RRMS, possibly by increasing activated Treg frequency. In addition, the NCM frequency and NCM/CM ratio had high predictive values for disease stability (AUC = 0.97 and 0.94, respectively), suggesting these markers are potential predictors of a long-term NEDA status in RRMS.
Collapse
Affiliation(s)
- Misako Minote
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Wakiro Sato
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
- Multiple Sclerosis Center, National Center of Neurology and Psychiatry, Kodaira, Japan
- Section of Research and Development Strategy, Translational Medical Center, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Kimitoshi Kimura
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Atsuko Kimura
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Youwei Lin
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
- Multiple Sclerosis Center, National Center of Neurology and Psychiatry, Kodaira, Japan
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Tomoko Okamoto
- Multiple Sclerosis Center, National Center of Neurology and Psychiatry, Kodaira, Japan
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Yamamura
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
- Multiple Sclerosis Center, National Center of Neurology and Psychiatry, Kodaira, Japan
| |
Collapse
|
38
|
Liu Y, Weng L, Wang Y, Zhang J, Wu Q, Zhao P, Shi Y, Wang P, Fang L. Deciphering the role of CD47 in cancer immunotherapy. J Adv Res 2024; 63:129-158. [PMID: 39167629 PMCID: PMC11380025 DOI: 10.1016/j.jare.2023.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 10/05/2023] [Accepted: 10/18/2023] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Immunotherapy has emerged as a novel strategy for cancer treatment following surgery, radiotherapy, and chemotherapy. Immune checkpoint blockade and Chimeric antigen receptor (CAR)-T cell therapies have been successful in clinical trials. Cancer cells evade immune surveillance by hijacking inhibitory pathways via overexpression of checkpoint genes. The Cluster of Differentiation 47 (CD47) has emerged as a crucial checkpoint for cancer immunotherapy by working as a "don't eat me" signal and suppressing innate immune signaling. Furthermore, CD47 is highly expressed in many cancer types to protect cancer cells from phagocytosis via binding to SIRPα on phagocytes. Targeting CD47 by either interrupting the CD47-SIRPα axis or combing with other therapies has been demonstrated as an encouraging therapeutic strategy in cancer immunotherapy. Antibodies and small molecules that target CD47 have been explored in pre- and clinical trials. However, formidable challenges such as the anemia and palate aggregation cannot be avoided because of the wide presentation of CD47 on erythrocytes. AIM OF VIEW This review summarizes the current knowledge on the regulation and function of CD47, and provides a new perspective for immunotherapy targeting CD47. It also highlights the clinical progress of targeting CD47 and discusses challenges and potential strategies. KEY SCIENTIFIC CONCEPTS OF REVIEW This review provides a comprehensive understanding of targeting CD47 in cancer immunotherapy, it also augments the concept of combination immunotherapy strategies by employing both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Yu'e Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Linjun Weng
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yanjin Wang
- Department of Nephrology, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Jin Zhang
- Department of Pharmacology and Toxicology, University of Mississippi, Medical Center, 39216 Jackson, MS, USA
| | - Qi Wu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Pengcheng Zhao
- School of Life Sciences and Medicine, Shandong University of Technology, No.266 Xincun West Road, Zibo 255000, Shandong Province, China
| | - Yufeng Shi
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China; Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai 200092, China.
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Lan Fang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
39
|
Mildner A, Kim KW, Yona S. Unravelling monocyte functions: from the guardians of health to the regulators of disease. DISCOVERY IMMUNOLOGY 2024; 3:kyae014. [PMID: 39430099 PMCID: PMC11486918 DOI: 10.1093/discim/kyae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 10/22/2024]
Abstract
Monocytes are a key component of the innate immune system. They undergo intricate developmental processes within the bone marrow, leading to diverse monocyte subsets in the circulation. In a state of healthy homeostasis, monocytes are continuously released into the bloodstream, destined to repopulate specific tissue-resident macrophage pools where they fulfil tissue-specific functions. However, under pathological conditions monocytes adopt various phenotypes to resolve inflammation and return to a healthy physiological state. This review explores the nuanced developmental pathways and functional roles that monocytes perform, shedding light on their significance in both physiological and pathological contexts.
Collapse
Affiliation(s)
- Alexander Mildner
- MediCity Research Laboratory, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, 20014 Turku, Finland
| | - Ki-Wook Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Simon Yona
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| |
Collapse
|
40
|
Bashore AC, Xue C, Kim E, Yan H, Zhu LY, Pan H, Kissner M, Ross LS, Zhang H, Li M, Reilly MP. Monocyte Single-Cell Multimodal Profiling in Cardiovascular Disease Risk States. Circ Res 2024; 135:685-700. [PMID: 39105287 PMCID: PMC11430373 DOI: 10.1161/circresaha.124.324457] [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: 02/17/2024] [Revised: 07/11/2024] [Accepted: 07/28/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Monocytes are a critical innate immune system cell type that serves homeostatic and immunoregulatory functions. They have been identified historically by the cell surface expression of CD14 and CD16. However, recent single-cell studies have revealed that they are much more heterogeneous than previously realized. METHODS We utilized cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) and single-cell RNA sequencing to describe the comprehensive transcriptional and phenotypic landscape of 437 126 monocytes. RESULTS This high-dimensional multimodal approach identified vast phenotypic diversity and functionally distinct subsets, including IFN-responsive, MHCIIhi (major histocompatibility complex class II), monocyte-platelet aggregates, as well as nonclassical, and several subpopulations of classical monocytes. Using flow cytometry, we validated the existence of MHCII+CD275+ MHCIIhi, CD42b+ monocyte-platelet aggregates, CD16+CD99- nonclassical monocytes, and CD99+ classical monocytes. Each subpopulation exhibited unique characteristics, developmental trajectories, transcriptional regulation, and tissue distribution. In addition, alterations associated with cardiovascular disease risk factors, including race, smoking, and hyperlipidemia were identified. Moreover, the effect of hyperlipidemia was recapitulated in mouse models of elevated cholesterol. CONCLUSIONS This integrative and cross-species comparative analysis provides a new perspective on the comparison of alterations in monocytes in pathological conditions and offers insights into monocyte-driven mechanisms in cardiovascular disease and the potential for monocyte subpopulation targeted therapies.
Collapse
Affiliation(s)
- Alexander C Bashore
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Chenyi Xue
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Eunyoung Kim
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Hanying Yan
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia (H.Y., M.L.)
| | - Lucie Y Zhu
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Huize Pan
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (H.P.)
| | - Michael Kissner
- Columbia Stem Cell Initiative, Department of Genetics and Development (M.K.), Columbia University Irving Medical Center, New York
| | - Leila S Ross
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Hanrui Zhang
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia (H.Y., M.L.)
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.)
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine (A.C.B., C.X., E.K., L.Y.Z., L.S.R., H.Z., M.P.R.), Columbia University Irving Medical Center, New York
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York (M.P.R.)
| |
Collapse
|
41
|
Hu Y, Tzeng SY, Cheng L, Lin J, Villabona-Rueda A, Yu S, Li S, Schneiderman Z, Zhu Y, Ma J, Wilson DR, Shannon SR, Warren T, Rui Y, Qiu C, Kavanagh EW, Luly KM, Zhang Y, Korinetz N, D’Alessio FR, Wang TH, Kokkoli E, Reddy SK, Luijten E, Green JJ, Mao HQ. Supramolecular assembly of polycation/mRNA nanoparticles and in vivo monocyte programming. Proc Natl Acad Sci U S A 2024; 121:e2400194121. [PMID: 39172792 PMCID: PMC11363337 DOI: 10.1073/pnas.2400194121] [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: 01/04/2024] [Accepted: 07/19/2024] [Indexed: 08/24/2024] Open
Abstract
Size-dependent phagocytosis is a well-characterized phenomenon in monocytes and macrophages. However, this size effect for preferential gene delivery to these important cell targets has not been fully exploited because commonly adopted stabilization methods for electrostatically complexed nucleic acid nanoparticles, such as PEGylation and charge repulsion, typically arrest the vehicle size below 200 nm. Here, we bridge the technical gap in scalable synthesis of larger submicron gene delivery vehicles by electrostatic self-assembly of charged nanoparticles, facilitated by a polymer structurally designed to modulate internanoparticle Coulombic and van der Waals forces. Specifically, our strategy permits controlled assembly of small poly(β-amino ester)/messenger ribonucleic acid (mRNA) nanoparticles into particles with a size that is kinetically tunable between 200 and 1,000 nm with high colloidal stability in physiological media. We found that assembled particles with an average size of 400 nm safely and most efficiently transfect monocytes following intravenous administration and mediate their differentiation into macrophages in the periphery. When a CpG adjuvant is co-loaded into the particles with an antigen mRNA, the monocytes differentiate into inflammatory dendritic cells and prime adaptive anticancer immunity in the tumor-draining lymph node. This platform technology offers a unique ligand-independent, particle-size-mediated strategy for preferential mRNA delivery and enables therapeutic paradigms via monocyte programming.
Collapse
Affiliation(s)
- Yizong Hu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Stephany Y. Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Leonardo Cheng
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Jinghan Lin
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Andres Villabona-Rueda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Shuai Yu
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL60208
| | - Sixuan Li
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Zachary Schneiderman
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Yining Zhu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Jingyao Ma
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| | - David R. Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Sydney R. Shannon
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Tiarra Warren
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Yuan Rui
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Chenhu Qiu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Erin W. Kavanagh
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Kathryn M. Luly
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Yicheng Zhang
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Nicole Korinetz
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Franco R. D’Alessio
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Tza-Huei Wang
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Efrosini Kokkoli
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Sashank K. Reddy
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Erik Luijten
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL60208
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL60208
- Department of Chemistry, Northwestern University, Evanston, IL60208
- Department of Physics and Astronomy, Northwestern University, Evanston, IL60208
| | - Jordan J. Green
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| |
Collapse
|
42
|
Noh JY, Han HW, Kim DM, Giles ED, Farnell YZ, Wright GA, Sun Y. Innate immunity in peripheral tissues is differentially impaired under normal and endotoxic conditions in aging. Front Immunol 2024; 15:1357444. [PMID: 39221237 PMCID: PMC11361940 DOI: 10.3389/fimmu.2024.1357444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Chronic low-grade inflammation is a hallmark of aging, aka "inflammaging", which is linked to a wide range of age-associated diseases. Immune dysfunction increases disease susceptibility, and increases morbidity and mortality of aging. Innate immune cells, including monocytes, macrophages and neutrophils, are the first responders of host defense and the key mediators of various metabolic and inflammatory insults. Currently, the understanding of innate immune programming in aging is largely fragmented. Here we investigated the phenotypic and functional properties of innate immune cells in various peripheral tissues of young and aged mice under normal and endotoxic conditions. Under the steady state, aged mice showed elevated pro-inflammatory monocytes/macrophages in peripheral blood, adipose tissue, liver, and colon. Under lipopolysaccharide (LPS)-induced inflammatory state, the innate immune cells of aged mice showed a different response to LPS stimulus than that of young mice. LPS-induced immune responses displayed differential profiles in different tissues and cell types. In the peripheral blood, when responding to LPS, the aged mice showed higher neutrophils, but lower pro-inflammatory monocytes than that in young mice. In the peritoneal fluid, while young mice exhibited significantly elevated pro-inflammatory neutrophils and macrophages in response to LPS, aged mice exhibited decreased pro-inflammatory neutrophils and variable cytokine responses in macrophages. In the adipose tissue, LPS induced less infiltrated neutrophils but more infiltrated macrophages in old mice than young mice. In the liver, aged mice showed a more robust increase of pro-inflammatory macrophages compared to that in young mice under LPS stimulation. In colon, macrophages showed relatively mild response to LPS in both young and old mice. We have further tested bone-marrow derived macrophages (BMDM) from young and aged mice, we found that BMDM from aged mice have impaired polarization, displaying higher expression of pro-inflammatory markers than those from young mice. These data collectively suggest that innate immunity in peripheral tissues is impaired in aging, and the dysregulation of immunity is tissue- and cell-dependent. Our findings in the rodent model underscore the complexity of aging immunity. Further investigation is needed to determine whether the immune profile observed in aged mice is applicable in age-associated diseases in humans.
Collapse
Affiliation(s)
- Ji Yeon Noh
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Hye Won Han
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Da Mi Kim
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Erin D. Giles
- School of Kinesiology, University of Michigan, Ann Arbor, MI, United States
| | - Yuhua Z. Farnell
- Department of Poultry Science, Texas A&M University, College Station, TX, United States
| | - Gus A. Wright
- Department of Veterinary Pathobiology, Texas A&M University,
College Station, TX, United States
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, United States
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, United States
| |
Collapse
|
43
|
Chen M, Wu Q, Shao N, Lai X, Lin H, Chen M, Wu Y, Chen J, Lin Q, Huang J, Chen X, Yan W, Chen S, Li H, Wu D, Yang M, Deng C. The significance of CD16+ monocytes in the occurrence and development of chronic thromboembolic pulmonary hypertension: insights from single-cell RNA sequencing. Front Immunol 2024; 15:1446710. [PMID: 39192976 PMCID: PMC11347785 DOI: 10.3389/fimmu.2024.1446710] [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: 06/10/2024] [Accepted: 07/17/2024] [Indexed: 08/29/2024] Open
Abstract
Background Chronic thromboembolic pulmonary hypertension (CTEPH) is a serious pulmonary vascular disease characterized by residual thrombi in the pulmonary arteries and distal pulmonary microvascular remodeling. The pathogenesis of CTEPH remains unclear, but many factors such as inflammation, immunity, coagulation and angiogenesis may be involved. Monocytes are important immune cells that can differentiate into macrophages and dendritic cells and play an important role in thrombus formation. However, the distribution, gene expression profile and differentiation trajectory of monocyte subsets in CTEPH patients have not been systematically studied. This study aims to reveal the characteristics and functions of monocytes in CTEPH patients using single-cell sequencing technology, and to provide new insights for the diagnosis and treatment of CTEPH. Methods Single-cell RNA sequencing (scRNA-seq) were performed to analyze the transcriptomic features of peripheral blood mononuclear cells (PBMCs) from healthy controls, CTEPH patients and the tissues from CTEPH patients after the pulmonary endarterectomy (PEA). We established a CTEPH rat model with chronic pulmonary embolism caused by repeated injection of autologous thrombi through a central venous catheter, and used flow cytometry to detect the proportion changes of monocyte subsets in CTEPH patients and CTEPH rat model. We also observed the infiltration degree of macrophage subsets in thrombus tissue and their differentiation relationship with peripheral blood monocyte subsets by immunofluorescence staining. Results The results showed that the monocyte subsets in peripheral blood of CTEPH patients changed significantly, especially the proportion of CD16+ monocyte subset increased. This monocyte subset had unique functional features at the transcriptomic level, involving processes such as cell adhesion, T cell activation, coagulation response and platelet activation, which may play an important role in pulmonary artery thrombus formation and pulmonary artery intimal remodeling. In addition, we also found that the macrophage subsets in pulmonary endarterectomy tissue of CTEPH patients showed pro-inflammatory and lipid metabolism reprogramming features, which may be related to the persistence and insolubility of pulmonary artery thrombi and the development of pulmonary hypertension. Finally, we also observed that CD16+ monocyte subset in peripheral blood of CTEPH patients may be recruited to pulmonary artery intimal tissue and differentiate into macrophage subset with high expression of IL-1β, participating in disease progression. Conclusion CD16+ monocytes subset had significant gene expression changes in CTEPH patients, related to platelet activation, coagulation response and inflammatory response. And we also found that these cells could migrate to the thrombus and differentiate into macrophages with high expression of IL-1β involved in CTEPH disease progression. We believe that CD16+ monocytes are important participants in CTEPH and potential therapeutic targets.
Collapse
Affiliation(s)
- Maohe Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Qiuxia Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Nan Shao
- Division of Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xingyue Lai
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Huo Lin
- Department of Pulmonary and Critical Care Medicine, Shishi County Hospital, Shishi, China
| | - Min Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Yijing Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Jiafan Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Qinghuang Lin
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Jiahui Huang
- Department of Respiratory and Critical Care Medicine, Fuqing City Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Xiaoyun Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Wei Yan
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Shi Chen
- Department of Respiratory and Critical Care, Wuhan No. 6 Hospital, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Hongli Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
- Department of Respiratory and Critical Care Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Dawen Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
- Department of Respiratory and Critical Care Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Minxia Yang
- Division of Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Chaosheng Deng
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
- Department of Respiratory and Critical Care Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
44
|
Li G, Yang H, Zhang D, Zhang Y, Liu B, Wang Y, Zhou H, Xu ZX, Wang Y. The role of macrophages in fibrosis of chronic kidney disease. Biomed Pharmacother 2024; 177:117079. [PMID: 38968801 DOI: 10.1016/j.biopha.2024.117079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/23/2024] [Accepted: 06/29/2024] [Indexed: 07/07/2024] Open
Abstract
Macrophages are widely distributed throughout various tissues of the body, and mounting evidence suggests their involvement in regulating the tissue microenvironment, thereby influencing disease onset and progression through direct or indirect actions. In chronic kidney disease (CKD), disturbances in renal functional homeostasis lead to inflammatory cell infiltration, tubular expansion, glomerular atrophy, and subsequent renal fibrosis. Macrophages play a pivotal role in this pathological process. Therefore, understanding their role is imperative for investigating CKD progression, mitigating its advancement, and offering novel research perspectives for fibrosis treatment from an immunological standpoint. This review primarily delves into the intrinsic characteristics of macrophages, their origins, diverse subtypes, and their associations with renal fibrosis. Particular emphasis is placed on the transition between M1 and M2 phenotypes. In late-stage CKD, there is a shift from the M1 to the M2 phenotype, accompanied by an increased prevalence of M2 macrophages. This transition is governed by the activation of the TGF-β1/SMAD3 and JAK/STAT pathways, which facilitate macrophage-to-myofibroblast transition (MMT). The tyrosine kinase Src is involved in both signaling cascades. By thoroughly elucidating macrophage functions and comprehending the modes and molecular mechanisms of macrophage-fibroblast interaction in the kidney, novel, tailored therapeutic strategies for preventing or attenuating the progression of CKD can be developed.
Collapse
Affiliation(s)
- Guangtao Li
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Hongxia Yang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Dan Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Zhi-Xiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| |
Collapse
|
45
|
Burgess MO, Janas P, Berry K, Mayr H, Mack M, Jenkins SJ, Bain CC, McSorley HJ, Schwarze J. Helminth induced monocytosis conveys protection from respiratory syncytial virus infection in mice. Allergy 2024; 79:2157-2172. [PMID: 38924546 DOI: 10.1111/all.16206] [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: 05/10/2023] [Revised: 04/17/2024] [Accepted: 05/08/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection in infants is a major cause of viral bronchiolitis and hospitalisation. We have previously shown in a murine model that ongoing infection with the gut helminth Heligmosomoides polygyrus protects against RSV infection through type I interferon (IFN-I) dependent reduction of viral load. Yet, the cellular basis for this protection has remained elusive. Given that recruitment of mononuclear phagocytes to the lung is critical for early RSV infection control, we assessed their role in this coinfection model. METHODS Mice were infected by oral gavage with H. polygyrus. Myeloid immune cell populations were assessed by flow cytometry in lung, blood and bone marrow throughout infection and after secondary infection with RSV. Monocyte numbers were depleted by anti-CCR2 antibody or increased by intravenous transfer of enriched monocytes. RESULTS H. polygyrus infection induces bone marrow monopoiesis, increasing circulatory monocytes and lung mononuclear phagocytes in a IFN-I signalling dependent manner. This expansion causes enhanced lung mononuclear phagocyte counts early in RSV infection that may contribute to the reduction of RSV load. Depletion or supplementation of circulatory monocytes prior to RSV infection confirms that these are both necessary and sufficient for helminth induced antiviral protection. CONCLUSIONS H. polygyrus infection induces systemic monocytosis contributing to elevated mononuclear phagocyte numbers in the lung. These cells are central to an anti-viral effect that reduces the peak viral load in RSV infection. Treatments to promote or modulate these cells may provide novel paths to control RSV infection in high risk individuals.
Collapse
Affiliation(s)
- Matthew O Burgess
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Piotr Janas
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Karla Berry
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Hannah Mayr
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Stephen J Jenkins
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Calum C Bain
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Henry J McSorley
- Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Jurgen Schwarze
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- Child Life and Health, Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
46
|
Lin J, Bai S, He L, Yang Y, Li X, Luo L, Wang Y, Chen YY, Qin J, Zhong Y. Cytotoxic Lymphocyte-Monocyte Complex Reflects the Dynamics of Coronavirus Disease 2019 Systemic Immune Response. J Infect Dis 2024; 230:5-14. [PMID: 39052699 DOI: 10.1093/infdis/jiae048] [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: 07/16/2023] [Revised: 12/01/2023] [Accepted: 01/29/2024] [Indexed: 02/03/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes a variety of clinical manifestations, many of which originate from altered immune responses, either locally or systemically. Immune cell cross-talk occurs mainly in lymphoid organs. However, systemic cell interaction specific to coronavirus disease 2019 has not been well characterized. Here, by employing single-cell RNA sequencing and imaging flow cytometry analysis, we unraveled, in peripheral blood, a heterogeneous group of cell complexes formed by the adherence of CD14+ monocytes to different cytotoxic lymphocytes, including SARS-CoV-2-specific CD8+ T cells, γδ T cells, and natural killer T cells. These lymphocytes attached to CD14+ monocytes that showed enhanced inflammasome activation and pyroptosis-induced cell death in progression stage; in contrast, in the convalescent phase, CD14+ monocytes with elevated antigen presentation potential were targeted by cytotoxic lymphocytes, thereby restricting the excessive immune activation. Collectively, our study reports previously unrecognized cell-cell interplay in the SARS-CoV-2-specific immune response, providing new insight into the intricacy of dynamic immune cell interaction representing antiviral defense.
Collapse
Affiliation(s)
- Jiajia Lin
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine
- Shanghai Immune Therapy Institute, Renji Hospital and Baoshan Branch of Renji Hospital, Shanghai Jiao Tong University School of Medicine
| | - Shiyu Bai
- Shanghai Immune Therapy Institute, Renji Hospital and Baoshan Branch of Renji Hospital, Shanghai Jiao Tong University School of Medicine
| | - Liheng He
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine
| | - Ye Yang
- Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| | - Xiyue Li
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine
- Shanghai Immune Therapy Institute, Renji Hospital and Baoshan Branch of Renji Hospital, Shanghai Jiao Tong University School of Medicine
| | - Liulin Luo
- Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine
| | - Ying Wang
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine
- Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying-Ying Chen
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine
- Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinhong Qin
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine
| | - Yi Zhong
- Shanghai Immune Therapy Institute, Renji Hospital and Baoshan Branch of Renji Hospital, Shanghai Jiao Tong University School of Medicine
| |
Collapse
|
47
|
Chang LA, Schotsaert M. Ally, adversary, or arbitrator? The context-dependent role of eosinophils in vaccination for respiratory viruses and subsequent breakthrough infections. J Leukoc Biol 2024; 116:224-243. [PMID: 38289826 PMCID: PMC11288382 DOI: 10.1093/jleuko/qiae010] [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: 10/29/2023] [Revised: 12/12/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
Eosinophils are a critical type of immune cell and central players in type 2 immunity. Existing literature suggests that eosinophils also can play a role in host antiviral responses, typically type 1 immune events, against multiple respiratory viruses, both directly through release of antiviral mediators and indirectly through activation of other effector cell types. One way to prime host immune responses toward effective antiviral responses is through vaccination, where typically a type 1-skewed immunity is desirable in the context of intracellular pathogens like respiratory viruses. In the realm of breakthrough respiratory viral infection in vaccinated hosts, an event in which virus can still establish productive infection despite preexisting immunity, eosinophils are most prominently known for their link to vaccine-associated enhanced respiratory disease upon natural respiratory syncytial virus infection. This was observed in a pediatric cohort during the 1960s following vaccination with formalin-inactivated respiratory syncytial virus. More recent research has unveiled additional roles of the eosinophil in respiratory viral infection and breakthrough infection. The specific contribution of eosinophils to the quality of vaccine responses, vaccine efficacy, and antiviral responses to infection in vaccinated hosts remains largely unexplored, especially regarding their potential roles in protection. On the basis of current findings, we will speculate upon the suggested function of eosinophils and consider the many potential ways by which eosinophils may exert protective and pathological effects in breakthrough infections. We will also discuss how to balance vaccine efficacy with eosinophil-related risks, as well as the use of eosinophils and their products as potential biomarkers of vaccine efficacy or adverse events.
Collapse
Affiliation(s)
- Lauren A Chang
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, United States
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1630, New York, NY 10029, United States
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States
| |
Collapse
|
48
|
Yang J, Chen S, Ma F, Ding N, Mi S, Zhao Q, Xing Y, Yang T, Xing K, Yu Y, Wang C. Pathogen stimulations and immune cells synergistically affect the gene expression profile characteristics of porcine peripheral blood mononuclear cells. BMC Genomics 2024; 25:719. [PMID: 39054472 PMCID: PMC11270792 DOI: 10.1186/s12864-024-10603-9] [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: 02/07/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Pigs serve as a crucial source of protein in the human diet and play a fundamental role in ensuring food security. However, infectious diseases caused by bacteria or viruses are a major threat to effective global pig farming, jeopardizing human health. Peripheral blood mononuclear cells (PBMCs) are a mixture of immune cells that play crucial roles in immunity and disease resistance in pigs. Previous studies on the gene expression regulation patterns of PBMCs have concentrated on a single immune stimulus or immune cell subpopulation, which has limited our comprehensive understanding of the mechanisms of the pig immune response. RESULTS Here, we integrated and re-analyzed RNA-seq data published online for porcine PBMC stimulated by lipopolysaccharide (LPS), polyinosinic acid (PolyI:C), and various unknown microorganisms (EM). The results revealed that gene expression and its functional characterization are highly specific to the pathogen, identifying 603, 254, and 882 pathogen-specific genes and 38 shared genes, respectively. Notably, LPS and PolyI:C stimulation directly triggered inflammatory and immune-response pathways, while exposure to mixed microbes (EM) enhanced metabolic processes. These pathogen-specific genes were enriched in immune trait-associated quantitative trait loci (QTL) and eGenes in porcine immune tissues and were implicated in specific cell types. Furthermore, we discussed the roles of eQTLs rs3473322705 and rs1109431654 in regulating pathogen- and cell-specific genes CD300A and CD93, using cellular experiments. Additionally, by integrating genome-wide association studies datasets from 33 complex traits and diseases in humans, we found that pathogen-specific genes were significantly enriched for immune traits and metabolic diseases. CONCLUSIONS We systematically analyzed the gene expression profiles of the three stimulations and demonstrated pathogen-specific and cell-specific gene regulation across different stimulations in porcine PBMCs. These findings enhance our understanding of shared and distinct regulatory mechanisms of genetic variants in pig immune traits.
Collapse
Affiliation(s)
- Jinyan Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technologyn, China Agricultural University, Beijing, 100193, China
| | - Siqian Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technologyn, China Agricultural University, Beijing, 100193, China
| | - Fuping Ma
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technologyn, China Agricultural University, Beijing, 100193, China
| | - Ning Ding
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technologyn, China Agricultural University, Beijing, 100193, China
| | - Siyuan Mi
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technologyn, China Agricultural University, Beijing, 100193, China
| | - Qingyao Zhao
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technologyn, China Agricultural University, Beijing, 100193, China
| | - Yue Xing
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technologyn, China Agricultural University, Beijing, 100193, China
| | - Ting Yang
- Dabei-Nong Science and Technology Group Co., Ltd, Beijing, 100080, China
| | - Kai Xing
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technologyn, China Agricultural University, Beijing, 100193, China
| | - Ying Yu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technologyn, China Agricultural University, Beijing, 100193, China.
| | - Chuduan Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technologyn, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
49
|
Zhu Z, Wang M, Lu S, Dai S, Liu J. Role of macrophage polarization in heart failure and traditional Chinese medicine treatment. Front Pharmacol 2024; 15:1434654. [PMID: 39104386 PMCID: PMC11298811 DOI: 10.3389/fphar.2024.1434654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Heart failure (HF) has a severe impact on public health development due to high morbidity and mortality and is associated with imbalances in cardiac immunoregulation. Macrophages, a major cell population involved in cardiac immune response and inflammation, are highly heterogeneous and polarized into M1 and M2 types depending on the microenvironment. M1 macrophage releases inflammatory factors and chemokines to activate the immune response and remove harmful substances, while M2 macrophage releases anti-inflammatory factors to inhibit the overactive immune response and promote tissue repair. M1 and M2 restrict each other to maintain cardiac homeostasis. The dynamic balance of M1 and M2 is closely related to the Traditional Chinese Medicine (TCM) yin-yang theory, and the imbalance of yin and yang will result in a pathological state of the organism. Studies have confirmed that TCM produces positive effects on HF by regulating macrophage polarization. This review describes the critical role of macrophage polarization in inflammation, fibrosis, angiogenesis and electrophysiology in the course of HF, as well as the potential mechanism of TCM regulation of macrophage polarization in preventing and treating HF, thereby providing new ideas for clinical treatment and scientific research design of HF.
Collapse
Affiliation(s)
- Zheqin Zhu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Min Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Shenghua Lu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Sisi Dai
- Hunan University of Chinese Medicine, Changsha, China
| | - Jianhe Liu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
50
|
Ding T, Wang Y, Meng Y, Wu E, Shao Q, Lin S, Yu Y, Qian J, He Q, Zhang J, Wang J, Kohane DS, Zhan C. Reciprocal Interaction with Neutrophils Facilitates Cutaneous Accumulation of Liposomes. ACS NANO 2024; 18:18769-18784. [PMID: 38950189 DOI: 10.1021/acsnano.4c06638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Liposomes are versatile drug delivery systems in clinical use for cancer and many other diseases. Unfortunately, PEGylated liposomal doxorubicin (sLip/DOX) exhibits serious dose-limiting cutaneous toxicities, which are closely related to the extravascular accumulation of sLip/DOX in the dermis. No clinical interventions have been proposed for cutaneous toxicities due to the elusive transport pathways. Herein, we showed that the reciprocal interaction between liposomes and neutrophils played pivotal roles in liposome extravasation into the dermis. Neutrophils captured liposomes via the complement receptor 3 (CD11b/CD18) recognizing the fragment of complement component C3 (iC3b) deposited on the liposomal surface. Uptake of liposomes also activated neutrophils to induce CD11b upregulation and enhanced the ability of neutrophils to migrate outside the capillaries. Furthermore, inhibition of complement activation either by CRIg-L-FH (a C3b/iC3b targeted complement inhibitor) or blocking the phosphate negative charge in mPEG-DSPE could significantly reduce liposome uptake by neutrophils and alleviate the cutaneous accumulation of liposomes. These results validated the liposome extravasation pathway mediated by neutrophils and provided potential solutions to the devastating cutaneous toxicities occurring during sLip/DOX treatment.
Collapse
Affiliation(s)
- Tianhao Ding
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yang Wang
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanchun Meng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
| | - Ercan Wu
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Qianwen Shao
- School of Pharmacy & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P. R. China
| | - Shiqi Lin
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yifei Yu
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Jun Qian
- School of Pharmacy & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P. R. China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
| | - Jing Wang
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
- School of Pharmacy & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P. R. China
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|